Anda di halaman 1dari 591

Fourth Edition

Nieschlag, Behre
Professor Eberhard Nieschlag Now in its fourth edition, Testosterone: Action, Deficiency,
is Emeritus and former Director Substitution provides the most up-to-date and comprehen-

and Nieschlag
of the Centre for Reproductive
Medicine and Andrology at the sive source of information on testosterone and its role in
University of Mnster, Germany. human physiology and pathology. It covers biosynthesis and

Testosterone
mechanisms of action, and reviews their effects on brain and
Professor Hermann M. Behre
behaviour, spermatogenesis, hair growth, bones, muscles,
is Director of the Center for
Reproductive Medicine and erythropoiesis, the cardiovascular system and the prostate.
Andrology of the University The therapeutic uses of testosterone preparations are care-
of Halle, Germany. fully evaluated as well as its abuse and detection in competi-
Action | Deficiency | Substitution

Testosterone
Fourth Edition
tive sport. This new edition also devotes sections to explor-
ing the effects of testosterone on female physiology and
sexual function, and the experimental use of testosterone in
male hormonal contraception. Edited by
Testosterone: Action, Deficiency, Substitution has been
written and edited by world experts in the fields of clinical
Eberhard Nieschlag
andrology and endocrinology, and provides a unique and Hermann M. Behre
invaluable guide for andrologists, endocrinologists, urolo-
Associate Editor
gists, internists and those working in reproductive medicine.
Susan Nieschlag

Other titles of interest:


The Sperm Cell
Edited by Christopher J. De Jonge and Christopher Barratt
(ISBN 9780521184175)
Fertility Preservation in Male Cancer Patients
Edited by John P. Mulhall in association with Linda D.
Applegarth, Robert D. Oates and Peter N. Schlegel
(ISBN 9781107012127)
In-Vitro Fertilization
Kay Elder and Brian Dale
(ISBN 9780521730723)

C O V E R D E S I G N : S U E WAT S O N

NIESCHLAG: TESTOSTERONE PPC CMYBLK


Testosterone
Action, Deficiency, Substitution
Fourth Edition
Testosterone
Action, Deficiency, Substitution
Fourth Edition
Edited by
Eberhard Nieschlag
Emeritus Professor and former Director, Center for Reproductive Medicine and Andrology, University of Mnster, Germany

Hermann M. Behre
Director, Center for Reproductive Medicine and Andrology of the University, Halle, Germany

Associate Editor
Susan Nieschlag
Center for Reproductive Medicine and Andrology, University of Mnster, Germany
CAMBRIDGE UNIVERSITY PRESS
Cambridge, New York, Melbourne, Madrid, Cape Town,
Singapore, So Paulo, Delhi, Mexico City

Cambridge University Press


The Edinburgh Building, Cambridge CB2 8RU, UK

Published in the United States of America


by Cambridge University Press, New York

www.cambridge.org
Information on this title: www.cambridge.org/9781107012905

Cambridge University Press 2004, 2012

This publication is in copyright. Subject to statutory exception


and to the provisions of relevant collective licensing agreements,
no reproduction of any part may take place without
the written permission of Cambridge University Press.

Third edition published 2004


Fourth edition published 2012

Printed andiBoundiin the United Kingdom by i the MPG iBooksiGroup

A catalog record for this publication is available from the British Library

Library of Congress Cataloging-in-Publication Data


Testosterone : action, deficiency, substitution / edited by
Eberhard Nieschlag, Hermann M. Behre ; associate editor,
Susan Nieschlag. 4th ed.
p. ; cm.
Includes bibliographical references and index.
ISBN 978-1-107-01290-5 (Hardback)
I. Nieschlag, E. II. Behre, H. M. (Hermann M.), 1961
III. Nieschlag, S. (Susan)
[DNLM: 1. Testosteronetherapeutic use. 2. Testosteronephysi-
ology. WJ 875]
615.30 66dc23
2011049739

ISBN 978-1-107-01290-5 Hardback

Cambridge University Press has no responsibility for the persistence or


accuracy of URLs for external or third-party internet websites referred to
in this publication, and does not guarantee that any content on such
websites is, or will remain, accurate or appropriate.

Every effort has been made in preparing this book to provide accurate
and up-to-date information which is in accord with accepted standards
and practice at the time of publication. Although case histories are
drawn from actual cases, every effort has been made to disguise the
identities of the individuals involved. Nevertheless, the authors, editors,
and publishers can make no warranties that the information contained
herein is totally free from error, not least because clinical standards are
constantly changing through research and regulation. The authors,
editors, and publishers therefore disclaim all liability for direct or
consequential damages resulting from the use of material contained in
this book. Readers are strongly advised to pay careful attention to
information provided by the manufacturer of any drugs or equipment
that they plan to use.
Contents
List of contributors vii
Preface xi

1 The medical and cultural history of 11 Testosterone, obesity, diabetes and the
testosterone and the testes 1 metabolic syndrome 235
Eberhard Nieschlag and Susan Nieschlag T. Hugh Jones and Kevin S. Channer

2 Testosterone: biosynthesis, transport, 12 Testosterone and erection 251


metabolism and (non-genomic) actions 15 Mario Maggi and Hermann M. Behre
C. Marc Luetjens and Gerhard F. Weinbauer 13 Testosterone and the prostate 268
3 Pathophysiology of the androgen John T. Isaacs and Samuel R. Denmeade
receptor 33 14 Clinical use of testosterone in hypogonadism
Olaf Hiort, Ralf Werner, and Michael Zitzmann and other conditions 292
4 Methodology for measuring testosterone, Eberhard Nieschlag and Hermann M. Behre
dihydrotestosterone and sex hormone- 15 Testosterone preparations for clinical use
binding globulin in a clinical setting 60 in males 309
Manuela Simoni, Flaminia Fanelli, Laura Roli, Hermann M. Behre and Eberhard Nieschlag
and Uberto Pagotto
16 Androgens in male senescence 336
5 The behavioral correlates of testosterone 87 Jean-Marc Kaufman, Guy TSjoen, and
John Bancroft Alex Vermeulen
6 The role of testosterone in 17 Androgen therapy in non-gonadal disease 372
spermatogenesis 123 David J. Handelsman
Liza ODonnell and Robert I. McLachlan
18 Review of guidelines on diagnosis and
7 Androgens and hair: a biological paradox treatment of testosterone deficiency 408
with clinical consequences 154 Ronald S. Swerdloff and Christina C. L. Wang
Valerie Anne Randall
19 Pathophysiology of estrogen action in
8 Testosterone and bone 177 men 421
Dirk Vanderschueren, Mieke Sinnesael, Vincenzo Rochira, Daniele Santi, and
Evelien Gielen, Frank Claessens, and Cesare Carani
Steven Boonen
20 Dehydroepiandrosterone and
9 Androgen effects on the skeletal muscle 191 androstenedione 437
Shalender Bhasin, Ravi Jasuja, Carlo Serra, Bruno Allolio, Wiebke Arlt, and Stefanie Hahner
Rajan Singh, Thomas W. Storer, Wen Guo,
Thomas G. Travison, and Shezad Basaria 21 The state-of-the-art in the development of
selective androgen receptor modulators 459
10 Testosterone and cardiovascular disease 207 Ravi Jasuja, Mikhail N. Zacharov, and
Kevin S. Channer and T. Hugh Jones Shalender Bhasin

v
Contents

22 The essential role of testosterone in hormonal 25 Sequelae of doping with anabolic


male contraception 470 steroids 535
Eberhard Nieschlag and Hermann M. Behre Elena Vorona and Eberhard Nieschlag
23 Testosterone use in women 494
Susan R. Davis
24 Detection of illegal use of androgens and Index 547
selective androgen receptor modulators 517 Color plate section between pages 244 and 245
Wilhelm Schnzer and Mario Thevis

vi
Contributors

Bruno Allolio Frank Claessens


Department of Medicine I, University of Wrzburg, Laboratory of Molecular Endocrinology,
Wrzburg, Germany Department of Cellular and Molecular Medicine,
KU Leuven, Leuven, Belgium
Wiebke Arlt
Division of Medical Sciences, Endocrinology Susan R. Davis
and Metabolism, University of Birmingham, Womens Health Research Program, School of Public
Birmingham, UK Health and Preventive Medicine, Department of
Epidemiology and Preventive Medicine, Monash
John Bancroft University, Melbourne, Victoria, Australia
Kinsey Institute for Research in Sex, Gender,
and Reproduction, Indiana University, Samuel R. Denmeade
Bloomington, IN, USA Johns Hopkins University, Department of Oncology,
Baltimore, MD, USA
Shezad Basaria
Flaminia Fanelli
Boston University School of Medicine,
Section of Endocrinology, Diabetes and Nutrition, Endocrinology Unit, S. Orsola-Malpighi Hospital,
Boston Medical Center, Boston, MA, USA Alma Mater University of Bologna, Bologna, Italy

Evelien Gielen
Hermann M. Behre
Laboratory of Molecular Endocrinology, Department
Center for Reproductive Medicine and Andrology
of Cellular and Molecular Medicine, KU Leuven,
of the University, Halle, Germany
Leuven, Belgium
Shalender Bhasin Wen Guo
Boston University School of Medicine, Section Boston University School of Medicine,
of Endocrinology, Diabetes and Nutrition, Section of Endocrinology, Diabetes and Nutrition,
Boston Medical Center, Boston, MA, USA Boston Medical Center, Boston, MA, USA
Steven Boonen Stefanie Hahner
Gerontology and Geriatrics Section, Department of Department of Medicine I, University of Wrzburg,
Clinical and Experimental Medicine, KU Leuven, Wrzburg, Germany
Leuven, Belgium
David J. Handelsman
Cesare Carani ANZAC Research Institute and Department of
Department of Biomedical, Metabolic and Neural Andrology, Concord Hospital, University of Sydney,
Sciences, University of Modena and Reggio Emilia, Sydney, NSW, Australia
Modena, Italy
Olaf Hiort
Kevin S. Channer Department of Pediatric Endocrinology and
Sheffield Hallam University and Royal Hallamshire Diabetology, Pediatric and Adolescent Medicine,
Hospital, Sheffield, UK University of Lbeck, Lbeck, Germany

vii
List of contributors

John T. Isaacs Vincenzo Rochira


The Johns Hopkins University, Oncology Center, Department of Biomedical, Metabolic and Neural
Baltimore, MD, USA Sciences, University of Modena and Reggio Emilia,
Modena, Italy
Ravi Jasuja
Boston University School of Medicine, Laura Roli
Section of Endocrinology, Diabetes and Nutrition, Department of Clinical Pathology,
Boston Medical Center, Boston, MA, USA Azienda USL of Modena, Modena, Italy
T. Hugh Jones Daniele Santi
Robert Hague Centre for Diabetes and Department of Biomedical,
Endocrinology, Barnsley Hospital NHS Foundation Metabolic and Neural Sciences,
Trust, Barnsley, and Department of Human University of Modena and Reggio Emilia,
Metabolism, University of Sheffield, Sheffield, UK Modena, Italy
Jean-Marc Kaufman
Wilhelm Schnzer
Department of Endocrinology, Ghent University
Institute of Biochemistry,
Hospital, Ghent, Belgium
Center for Preventive Doping Research,
C. Marc Luetjens German Sports University Cologne,
Division of Research and Safety Assessment, Cologne, Germany
Covance Laboratories GmbH, Mnster, Germany
Carlo Serra
Mario Maggi Boston University School of Medicine,
Unit di Andrologia, Dipartimento di Fisiopatologia Section of Endocrinology, Diabetes and Nutrition,
Clinica, Florence, Italy Boston Medical Center, Boston, MA, USA

Robert I. McLachlan Manuela Simoni


Prince Henrys Institute of Medical Research, Department of Biomedical, Metabolic and Neural
Clayton, Victoria, Australia Sciences, University of Modena and Reggio Emilia,
Modena, Italy
Eberhard Nieschlag
Center for Reproductive Medicine and Andrology, Rajan Singh
University of Mnster, Mnster, Germany Boston University School of Medicine,
Section of Endocrinology, Diabetes and Nutrition,
Susan Nieschlag
Boston Medical Center, Boston, MA, USA
Center for Reproductive Medicine and Andrology,
University of Mnster, Mnster, Germany Mieke Sinnesael
Liza ODonnell Clinical and Experimental Endocrinology Section,
Prince Henrys Institute of Medical Research, Department of Clinical and Experimental Medicine,
Clayton, Victoria, Australia KU Leuven, Leuven, Belgium

Uberto Pagotto Thomas W. Storer


Endocrinology Unit, S. Orsola-Malpighi Hospital, Boston University School of Medicine,
Alma Mater University of Bologna, Section of Endocrinology, Diabetes and Nutrition,
Bologna, Italy Boston Medical Center, Boston, MA, USA

Valerie Anne Randall Ronald S. Swerdloff


Department of Biomedical Sciences, Department of Medicine/Endocrinology,
University of Bradford, Bradford, UK Harbor-UCLA Medical Center, Torrance, CA, USA

viii
List of contributors

Mario Thevis Elena Vorona


Institute of Biochemistry, University Clinics of Mnster, Clinic for
Center for Preventive Doping Research, Transplantation Medicine, Mnster, Germany
German Sports University Cologne, Cologne,
Germany Christina C. L. Wang
UCLA Clinical and Translational Science Institute,
Thomas G. Travison Harbor-UCLA Medical Center and Los Angeles
Boston University School of Medicine, Biomedical Research Institute, Torrance, CA, USA
Section of Endocrinology, Diabetes and
Nutrition, Boston Medical Center, Gerhard F. Weinbauer
Boston, MA, USA Covance Laboratories GmbH, Mnster, Germany

Ralf Werner
Guy TSjoen
Division of Paediatric Endocrinology and Diabetes,
Department of Endocrinology, Ghent University
Department of Paediatrics, University of Lbeck,
Hospital, Ghent, Belgium
Lbeck, Germany
Dirk Vanderschueren Mikhail N. Zacharov
Clinical and Experimental Endocrinology Section, Boston University School of Medicine,
Department of Clinical and Experimental Medicine, Section of Endocrinology, Diabetes and Nutrition,
KU Leuven, Leuven, Belgium Boston Medical Center, Boston, MA, USA

Alex Vermeulen Michael Zitzmann


Department of Endocrinology, Ghent University Center for Reproductive Medicine and Andrology,
Hospital, Ghent, Belgium University of Mnster, Mnster, Germany

ix
Preface

Testosterone is the focal point of male reproductive scientist wishing for an authoritative overview of this
health. Testosterone is responsible for all male charac- central area of male reproductive health.
teristics, and testosterone deficiency results in hypo- In order to synchronize the writing of the various
gonadism which has an impact on the entire male chapters, the authors assembled at Castle Hohen-
organism. Testosterone plays an important role in kammer in Bavaria in October 2011 for three days,
male aging. Many diseases, especially mood disorders, and finalized their writings during that time. This
cardiovascular diseases and the metabolic syndrome served to unify the work as a whole, and ensures that
can be caused by testosterone deficiency, but can also it represents the most up-to-date reference source
result in hypogonadism. Consequently, testosterone is on testosterone and its many facets. This coordinated
a key factor in the treatment of male disorders; benefits effort of all contributors guarantees a long half-life
and risks of such treatment need to be balanced. The of this book as an up-to-date reference source.
potent effects of testosterone also give rise to their The editors wish to thank the authors of the vari-
misuse, prompting the establishment of sensitive ous chapters for their excellent compliance and timely
detection systems, but also bearing adverse side- submission of manuscripts. We could not have con-
effects. Measuring testosterone in body fluids based cluded this task without the tireless reading capacity
on immunoassays was the basis for research in male and help from Susan Nieschlag, who as a professional
pathophysiology over decades; however, methodology edited all manuscripts; nor would the volume have
is currently shifting to gas or liquid chromatography come to fruition without the skilful assistance of
tandem mass spectrometry. The hopes for a male hor- our secretaries Maria Schalkowski and Ina Nelles,
monal contraceptive rest on testosterone as its major who processed the manuscripts expediently. Finally,
constituent. As estrogens derived from testosterone the project and the meeting would not have been
play a role in male physiology, testosterone also is possible without the generous support from Bayer
of importance for general and sexual health in women. Health Care (formerly Bayer Schering Pharma), the
Finally, new therapeutic applications are expected from International Society of Andrology (ISA), the Euro-
selective androgen receptor modulators (SARMs). pean Academy of Andrology (EAA), the German
A tremendous amount of new knowledge has Association for Reproductive Health, the Clinical
accumulated over the eight years since the third Research Group for Reproductive Medicine, Galen
edition of this book on testosterone appeared. The 25 Pharma, Merck Serono, Ferring Pharmaceuticals
chapters of the current volume, written by worldwide and our home institutions, the Centers for Repro-
leading experts in their field, encapsulate this progress. ductive Medicine and Andrology at the Universities
The book appeals to the clinician as well as to the basic of Mnster and Halle.

xi
Chapter
The medical and cultural history

1 of testosterone and the testes


Eberhard Nieschlag and Susan Nieschlag

1.1 Introduction 1 1.3 Detours on the way to modern medicine 6


1.2 From antiquity to the age of 1.3.1 Organotherapy 6
enlightenment 2 1.3.2 Testis transplantations 7
1.2.1 Ancient Egypt 2 1.4 Twentieth-century science and medicine 8
1.2.2 Greco-Roman period 2 1.4.1 The rise of steroid biochemistry 8
1.2.3 Castration through the times 2 1.4.2 Testosterone is born 8
1.2.4 Power through polyorchidism 4 1.4.3 Early descriptions of syndromes of
1.2.5 First description of testicular morphology hypogonadism 10
and sperm 5 1.5 Key messages 10
1.2.6 Proof of endocrine function 5 1.6 References 11

1.1 Introduction science, these observations are often entertaining for


Although the term hormone was coined only as late as todays reader, but at the same time they are also
1905 (Starling 1905; Henderson 2005), the biological educational for the medical scholar. In addition, they
effects of hormones were known for much longer reflect the socio-cultural environment of the time when
and probably from the early beginnings of humankind. they were observed or exploited, so that the history of
The over- or underproduction of hormones as the fun- testosterone and the testes is not only of medical inter-
damental pathophysiological principle in endocrine dis- est, but also of interest to the general public.
orders often produces phenotypical alterations that can The authors of this chapter are not trained histor-
be easily recognized. Similarly, elimination of hormones ians, but the first author has a keen interest in the
by removing the respective gland or replacing the hor- history of medicine, as documented early by his first
mones in a deficient organism is a basic tool of experi- medical publication dealing with the biography of
mental endocrinology. In their exposed scrotal position, Otto Deiters (18341863), the describer of the Dei-
the testes are easily accessible for manipulation, be it by ters cells and nucleus (Nieschlag 1965). While spe-
accidental or by inflicted trauma. The causal relationship cializing in endocrinology and andrology, historical
between absence of the glands and the resulting bio- events surrounding testosterone were collected along
logical effects before or after puberty is easily recog- with the scientific studies, culminating in the request
nizable, and that is why effects of testosterone, or rather by the Royal College of Physicians (London) in 2005
its absence, have been known from early on. Victor to commemorate the hundredth anniversary of the
C. Medvei (1993), the master of the history of endocrin- term hormone by a talk on The history of testos-
ology, summarized this phenomenon by saying The terone, thus precipitating more systematic research
oldest key to the endocrine treasure trove: the testicles. on the subject. Since then a more complete but still
Indeed the lack of functional effects of the testes has patchy picture has been developed, which is presented
been noted since antiquity and, in the light of modern here. As it is impossible to give a full history of

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

1
Chapter 1: History of testosterone

testosterone and the testes here, and many develop- Aristotle also developed a theory of fertilization:
ments occurred simultaneously in various epochs, the semen is the activating agonist of the soul fertilizing
material is summarized under three more or less the passive female soil. Like his other work, this
systematic subheadings as follows (1.21.4). Never- theory had a long-lasting effect on science and was
theless, this is not a systematic review, but rather a only superseded by Leeuwenhoek and Spallanzani
collection of historical highlights. about 2000 years later (see below).
Also dating from the Hellenistic time (about 300
BC) until the first century AD, the fertility cult
1.2 From antiquity to the age around Diana/Artemis Ephesina was popular in the
of enlightenment entire Greco-Roman world, and was especially pro-
moted by the emperors Trajan (53117) and Hadrian
1.2.1 Ancient Egypt (76138). This cult had its origin in the temple of
One of the oldest hypogonadal patients whose image Artemis in Ephesos, the Artemision, where the
has been preserved appears to be Vizier Hemiunu, the magna mater was worshipped. The statues, of which
architect of the pyramid of Giza. The statue in his an excellent example can be seen in the National
mausoleum dating from 2531 BC (Roemer-Pelizaeus Archaeological Museum in Naples, are characterized
Museum, Hildesheim) shows a seated man with by multiple oval objects fixed to the goddesss frontal
straight hairline, no beard, bilateral gynecomastia thorax. Until quite recently these were interpreted as
and myxedematous lower legs. In contrast, other breasts, but were in reality bull or ram testes whose
statues from that period show well-built athletic bearers were sacrificed to the goddess, following
men; for instance the statue of Amenemhat II which their testes were fixed to the originally wooden
(19191884 BC) (which travelled in 2011 from the statue, then emanating fertility and potency to the
Egyptian Museum in Berlin to be exhibited in the worshipper. This cult is thus based on knowledge of
Metropolitan Museum in New York). The plaque on the powers of generation connected with the testes.
Hemiunus statue describes his achievements as an
architect, but does not mention a wife or children as
was customary at the time, indicating that he may 1.2.3 Castration through the times
have been infertile. He could have suffered from In Greek mythology castration already occurred
hypopituitarism including hypogonadism, perhaps among the first generation of gods. Gaea, mother
caused by a prolactinoma, but Klinefelter syndrome earth, grew out of the chaos and produced Uranos
would also be a possible diagnosis. by parthenogenesis, with whom she then generated
the titan Chronos. When Uranos prevented Gaea
from creating children with their son Chronos, she
1.2.2 Greco-Roman period induced Chronos to castrate his father. This episode
Aristotle (384322 BC), the universal genius and phil- has been depicted beautifully in a fresco by Giorgio
osopher of the Hellenistic era, observed and wrote on Vasari (15111574) in the Palazzo Vecchio in Flor-
The Generation of Animals, in which he described the ence. Uranos testes, thrown into the sea, caused the
generation of visible organs in fertilized chicken eggs. water to foam, and out of these bubbles the foam-
He knew the effects of castration in men as well as in born goddess of love Aphrodite (= Venus) was born.
animals, and described its consequences for animal Quite extraordinary events in terms of reproductive
husbandry. For humans, he developed the fascinating physiology!
hypothesis that the right testis produces boys and the In the real world, castration has been practiced
left testis girls. This could be based on case reports of for socio-cultural purposes since antiquity. Its major
men with unilateral anorchia or traumatic loss of one purpose was to generate obedient slaves who were
testis. To date no study could be traced verifying or loyal to their masters or rulers and who, being infer-
falsifying this hypothesis, and a proposed controlled tile, could not create competing offspring. Set to
clinical study to be performed today would most guarding harems, they also, and in larger numbers,
likely not be met by agreement of an ethics commit- obtained influential administrative and political pos-
tee. This also demonstrates how misleading case itions as in China, and formed elite troops (Mitamura
reports may be. 1992; Flaig 2009).

2
Chapter 1: History of testosterone

Although most likely already practiced earlier, the combined with blinding was administered for high
earliest documentation of creating eunuchs in China treason, especially when the insurgent was a close
dates back to about 1300 BC. The Chinese eunuch relative whom one did not want to kill directly. As
system, with several thousands at a time, continued told in the Islendinga Saga, Sturla of the Sturlungar
until the end of the imperial system in 1912. In the Clan in Iceland castrated and blinded his rebelling
nineteenth century there were still about 2000 relative Oraekja Snorrason in 1236 (personal commu-
eunuchs at the imperial court in Beijing. The last nication from U. Ebel, Chair of Scandinavian
Chinese eunuch died at the age of 93 in 1996. Only Sciences, University of Mnster, 2007). When the
the fact that imperial eunuchs could obtain high- Normans migrated south they also introduced this
ranking positions and considerable power as well as penal practice in the areas they invaded. When he
wealth makes it plausible that adult men underwent established his reign in Britain after 1066, William
this gruesome operation. It was performed by the Conqueror abolished the Anglo-Saxon death pen-
licensed surgeons just outside the imperial court alty and replaced it by castration and blinding: I also
in Beijing, by cutting off testes and penis. About forbid that anyone shall be slain or hanged for any
25% of the volunteers did not survive this bloody fault, but let his eyes be put out and let him be
operation. The severed genitals were kept in a box, castrated (Van Eickels 2004). As a further example,
as shown in the film The Last Emperor (1987), and in Sicily in 1194 King William III was castrated and
were eventually buried with their owner. blinded after a rebellion against Emperor Henry VI.
Eunuchs probably already existed in ancient This episode forms the historical background for
Egypt. From the times of the legendary Queen Klingsors castration in the Parsifal epos (Tuchel
Semiramis (about 800 BC) eunuchs were reported 1998). The Toulouse Law Codex of 1296 described
from Assyria, and the system developed and con- (and depicted) castration for high treason.
tinued into the Islamic world in the Middle East and Throughout the centuries, castration was applied
North Africa. Over centuries, slaves were deported to beaten enemies by victorious soldiers for revenge
from Sub-Saharan Africa to the Islamic cities and and as a measure to eliminate the enemies without
courts, and many of the slaves who survived the outright killing. When Italian troops invaded Ethiopia
exhausting march through the desert were then cas- and lost the battle of Aduwa in 1896, supposedly 7000
trated to serve as laborers, guards, administrators and Italian soldiers were castrated (Melicow 1977). As
even soldiers (Barth 1857). It is astonishing that these reported by Babtschenko (2007), this still happened
tasks could be fulfilled without the anabolic effects of on both sides during the Chechen War in the
testosterone. Caucasus in 1996.
It has been estimated that the transatlantic deport- Castration has also been reported as self-mutila-
ation of Africans to the Americas between 1450 and tion for religious reasons since ancient times in order
1870 comprised about 11.5 million people, while the to make a life of chastity easier. The early church
entire Islamic deportation of slaves from Africa father Origines (186254) is one of the most promin-
between 650 and 1920 amounted to 17 million people, ent examples. In the eleventh to fourteenth centuries
and several million of these African slaves were cas- the sect of the Catharers, with their strongholds in
trated. This constant drain of manpower effectively Southern France, promulgated self-castration as part
prevented economic and cultural development of of a pure life. More recently, castration was prac-
Sub-Saharan Africa. In medieval times slaves were ticed in Southern Russia among members of the
also exported from Europe to the Islamic countries. Scoptic sect founded in the eighteenth century, and
These slaves were mainly from Eastern European the medical consequences were documented (Wilson
(Slavic) and Central Asian countries. There were and Roehrborn 1999). The largest contemporary
well-established slave routes through Europe, and group of castrates are the hijras in India. They func-
Verdun has the questionable historical fame of having tion as professional well-wishers at birth rites, and
been the European center for castration of slaves on receive considerable financial rewards. Several thou-
their way from the East to the South at those times sand of them exist.
(Flaig 2009). Castration has also been used as revenge for
Castration has also been practiced as lawful pun- seduction and adultery through the centuries. For
ishment. In medieval Scandinavia, castration example, Paris has been reported to have castrated

3
Chapter 1: History of testosterone

Peritanos after he had seduced his famous wife 100


Helena (Lehrs 1832). The case of the great medieval 90
theologian and philosopher Peter Abaelard (1079 80
1142) has been celebrated in history and literature.
70
As master of the cathedral school in Paris he seduced
60
one of his disciples, Heloise (11001164), whose uncle
then had Abaelard castrated by paid criminals. Des- 50
pite the lack of testosterone, one of the most romantic 40
love stories documented by literature developed 30
(Podlech 1990). This type of revenge continues into 20
most recent times as demonstrated by an incident in p = 0.65 (non-significant)
10
Germany in 2011 when the father of a 17-year-old girl 0
castrated her 57-year-old lover (Holzhaider 2011). Castrates Intact singers
These people had migrated to Germany from Kazakh- Fig. 1.1 Longevity of intact and castrated singers (n = 50 in
stan and might have brought rules of self-justice with each group) born between 1580 and 1859 (matched pairs of
intact and castrated singers with similar birth dates were formed).
them. (Box and whisker plots: solid line = median; broken line = mean
Castration before puberty maintains the high value) (Nieschlag et al. 1993).
voice of boys, so that soprano and alto voices with
the acoustic volume of an adult male result. Such Prepubertal castration provides an involuntary
high-pitched voices were considered desirable experiment on the influence of testosterone on lon-
among music lovers, especially at times when gevity. A retrospective comparison of the life expect-
women were not allowed to sing in church or in ancy of singers born between 1580 and 1859 and
operatic performances. Prepubertal castrates castrated before puberty, in order to preserve their
belonged to casts of operas in the seventeenth and high voices, against intact singers born at the same
eighteenth centuries; in the Vatican choirs these time, did not reveal a significant difference between
voices could be heard until the early twentieth cen- the lifespan of intact and castrated singers (Fig.1.1;
tury. Some of these castrates became famous soloists, Nieschlag et al. 1993). This would imply that the
such as Carlo Farinelli (17051782) or Domenico presence or absence of normal male testosterone
Annibaldi (17051779) (Melicow 1983; Ortkemper levels has no influence on life expectancy.
1993; Jenkins 1998). The middle Italian city of
Nurcia was a center for the operation on young boys.
However, most of the thousands of prepubertal 1.2.4 Power through polyorchidism
castrates lost their virility in vain as they did not While removal of the testes was used to punish or to
achieve the promised career as a singer, developed weaken the enemy or to obtain specific effects as a
only mediocre voices and were ridiculed by their consequence of the lack of testosterone, a super-
contemporaries. An impression of the castrato voice, numerary testis was always considered a sign of extra-
although of very low recording quality, is preserved ordinary virility and sexual vigor. Several such cases
from the last Vatican castrato, Alessandro Moreschi have been reported anecdotally. The Venetian Con-
(18581922), in one of the earliest gramophone dottiero Bartolomeo Colleoni (14001475), whose
recordings, made in 1902 (available today on CD). success on the battlefield as well as in the bedroom
Today countertenors applying a trained falsetto sing was legion (FrigeniCareddu 1996), advertised his
the castrato roles in, for example, Hndel operas, but physical abnormality by showing three testes in his
their head voices probably only approximate those coat of arms (Colleoni in Italian means testes;
of seventeenth-century castrati. Another impression nowadays spelled coglioni).
of the enormous artistic talents of the castrati is Allegedly, in 1539 Philipp Magnanimus, Count of
provided by the recordings of the mezzo-soprano Hesse (15041567), a protagonist of the Protestant
Cecilia Bartoli, who trained her voice to sing the reformation, was granted permission by Martin
extremely demanding arias by Nicola Porpora Luther to have two wives simultaneously: Christine
(16861768), Georg Friedrich Hndel (16851759) von Sachsen (15051549) and Margarethe von der
and others (Bartoli 2009). Saale (15221566), because of his three testes.

4
Chapter 1: History of testosterone

Because at that time bigamy was forbidden Leeuwenhoek 1948). Considering the primitive
upon penalty of death, Philipp got in even more appearance of his microscope, the details of his mor-
trouble with the Holy Roman Emperor than his phological descriptions of sperm are amazing, and it
ambitions as a religious reformer had caused. is even more amazing that 300 years later we are still
A third testis had already been diagnosed in the quarreling about normal and abnormal sperm
boy, but without ultrasound, biopsy or post- morphology (Kremer 1979).
mortem examination it remains unclear what these But it took another century until Lazzaro Spallan-
third structures in his and other famous peoples zani (17291799), a priest and scientist in Modena,
scrotums were possibly real testes or large sper- artificially inseminated frogs and dogs and demon-
matoceles? Whatever it was, this diagnosis had a strated the real function of sperm (Spallanzani 1779).
tremendous impact on the person and on historical By using sperm that he had preserved on ice he also
events. became the father of cryopreservation, without which
modern reproductive medicine and medicine in gen-
1.2.5 First description of testicular eral would be unthinkable.
He was a very systemic investigator and insisted
morphology and sperm quite in contrast to others at the time that experi-
The declaration of the Netherlands as an independent ments needed to be repeated before results could be
state during the Thirty Years War (16181648), and accepted (Gaeto 1999), a principle that prevails until
legalized at the Westphalian Peace Treaty in Mnster today.
in 1648, resulted in an enormous upswing in econ- The anatomist Franz Leydig (18211908) in
omy, culture and science in this country. The medical Wrzburg described the interstitial cells of the testes
sciences also boomed, based on proper research, espe- in 1850 (Leydig 1850). Although he did not know
cially in anatomy, as shown in Rembrandts painting their function, they still carry his name (Christensen
Anatomy of Dr. Tulp (1632). 1996). Finally in Milan, in 1865 Enrico Sertoli (1842
The reproductive sciences benefited from this 1910) discovered the supporting cells in the semi-
boom as well. It was Regnier de Graaf (16411673) niferous tubules (Sertoli 1865), also carrying his name
who not only described the Graafian follicle (1672), to date (Virdis 2005).
but also published a book about the anatomy of the Thus, over roughly two centuries the basic mor-
male reproductive tract as well as the treatment of its phological elements of the testes had been described,
disorders (de Graaf 1668). He produced very detailed as well as the one major product of the testes, the
drawings and descriptions of the male genital organs, sperm. Even the function of sperm and fertilization
and was the first to discover that the testes were had been elucidated, so that the time had come to
composed of a collection of minute vessels or tubules explore the basis of testicular endocrine function.
which confect semen; if these tubules were disentan-
gled without being broken and tied to one another,
they would far exceed 20 Dutch ells in length. 1.2.6 Proof of endocrine function
Having first described this in the edible dormouse, Although the endocrine function of the testes was
he then went on to the human: a classical case of known through their physiological and clinical
translational medicine. Unfortunately, de Graaf effects, their nature remained completely obscure.
became involved in a quarrel with his contemporary, While William Harvey (15781657) had discovered
Jan Swammerdam (16371680), about the question of the role of the heart and blood circulation in 1628, in
who had first described the follicles, and during that some medical schools Galens (AD 129216 ) concept
phase he died under nebulous circumstances at the of the four bodily humors prevailed well into the
young age of 32 (Setchell 1974). nineteenth century. Against this background, it is
A few years after Regnier de Graafs early and not surprising that the idea of a hormone working
mysterious death, his friend Antoni A. Leeuwenhoek as a signal transduced by circulating blood took so
(16321723), together with the student Johan Hamm, long to be born.
used his newly invented prototype of a microscope John Hunter (17281793) is considered by some
and described the little animals of the sperm in a to be the father of endocrinology, as he transplanted
letter to the Royal Society in London in 1677 (van testes in chickens. However, his outstanding

5
Chapter 1: History of testosterone

achievement as a scientist notwithstanding, he 1.3 Detours on the way to modern


transplanted testes in order to demonstrate the
vital principle of living organs. As a surgeon in medicine
the Seven Years War (17561763), he saw the need 1.3.1 Organotherapy
for transplantation of organs and limbs, and this is
As it was known that removal of the testes caused the
what stimulated his research. He never described his
clinically evidenced symptoms of hypogonadism,
testis transplantations himself, but we learn about
including impotence, prescribing ingestion of testes
them through a scholar, Dr. W. Irvine, in a letter to
to remedy the symptoms was a medical reflex inher-
Professor Th. Hamilton in Glasgow in 1771:
ent in organotherapy, practiced since antiquity. Thus
. . .Nay more, he has many hens just now into
the Roman Gaius Plinius Secundus (AD 2379) rec-
whose abdomen when young, he has put the testes
ommended the consumption of animal testes to treat
of a cock just separated from his body and his testis
symptoms of testosterone deficiency. Slightly more
has got blood vessels and nerves from the part of the
refined was the prescription of testicular extracts for
abdomen or viscera to which it is applied. . . Far
the same purpose in Arabic medicine, for example by
from any endocrine thought, the goal was to dem-
Mensue the Elder (777837) in Baghdad. Also, in
onstrate the survival of the transplant (Barker
China, raw and desiccated testes were prescribed,
Jorgensen 1971).
documented at least in the twelfth century by Hsue
Such thoughts were precipitated by Arnold
Shu-Wei. Around the same time, Albertus Magnus
Adolph Bertholds (18031861) experiments, which
(11931280) in Cologne, better known as a philoso-
also concerned transplanting chicken testes. As pub-
pher, recommended powdered hog testes, but refined
lished in 1849, he castrated four cocks: two received
his recipe by offering the powder in wine (Medvei
an ectopic transplantation of one testis, the two others
1993).
remained untreated, and he observed They crowed
Since early documentation, these potions con-
quite considerably, often fought among themselves
tinued to be prescribed and consumed up into
and with other young roosters and showed a normal
the twentieth century. In the 1920s Testifortan
inclination to hens. . . Since the testes can no longer
became a financially successful drug for treatment of
remain in connection with their original nerves after
impotence (Hirschfeld 1927). Its main constituent
being transplanted to a strange place. . . it follows that
was testis extracts and yohimbine, and after the war
the consensus in question must be affected through
17a-methyltestosterone was added without changing
the productive relationship of the testes, that is to say,
the name. Another famous preparation from the 1920s
through their action on the blood, and then through
and still marketed today is Okasa which, among other
the suitable ensuing action of the blood on the organ-
components, also contains testis sicca and thereby
ism as a whole. (Berthold 1849). The paper describes
small amounts of testosterone, as we could determine
only four animals and comprises only four pages in
in the 1970s (unpublished data). However, as the testes
contrast to the extensive style of the time but was
synthesize testosterone but do not store it, the daily
epochal. However, Bertholds rival at the University of
production of an adult man of about 68 mg is
Gttingen, Rudolf Wagner (18051864) was jealous,
contained in roughly 1 kg of (bull) testes, and even if
tried to repeat the experiments, but failed and
this amount of testosterone were to be consumed, the
declared them as rubbish (Wagner 1852). And as he
testosterone taken orally would be inactivated by the
became the full professor of physiology, his opinion
first-pass effect in the liver (Nieschlag et al. 1977).
prevailed. Bertholds personality did not allow him to
Therefore, all testicular organ therapy administered
fight for recognition of his findings (Simmer and
orally can only be considered as a placebo medication,
Simmer 1961; Simmer 1980).
which, however, may not be without its own effects
As blood circulation, the essential morphological
(Bundesrztekammer 2011). Ultimately this type of
elements of the testes and the biological and clinical
testicular organotherapy was terminated by the advent
effects of their endocrine action were known, history
of phosphodiesterase inhibitors.
could have continued straight from here to the dis-
Organotherapy literally exploded at the end of the
covery of testosterone, but due to these interpersonal
nineteenth century when Charles E. Brown-Squard
quarrels and other unhappy events, further develop-
(18471894), who until then was a well-reputed
ments took an oblique route.

6
Chapter 1: History of testosterone

scientist and member of several scientific academies, Later on he turned to animals as sources for his tes-
published the results of his famous self-experimen- ticular grafts and reported satisfaction on the part of
tation in the Lancet (Brown-Squard 1889). He gave the patients including 13 physicians (Stanley 1923).
himself 1-ml injections of a mixture of one part These surgeons had followers in many parts of the
testicular vein blood, one part semen and one part world, for example even in Iceland where, in 1929, the
juice extracted from dog or guinea-pig testes daily, surgeon Jonas Sveinsson transplanted testis slices
and after 20 days made astonishing observations on from a poor farmer in need of money to a rich
himself: A radical change took place in me. . . I had Norwegian businessman who, he then claimed, satis-
regained at least all the strength I possessed a good fied his 23-year-old wife so that he even had three
many years ago. I was able to make experiments for children with her (Sveinsson 1969). In the Soviet
several hours. After dinner I was able to write a paper Union, experimentation with human testicular
on a difficult subject. My limbs, tested with a dyna- transplantation continued at least into the 1980s
mometer, gained 6 to 7 kg in strength. The jet of urine (Shumankov and Gotsiridze 1978). The only testicular
and the power of defecation became stronger. transplantation resulting in fertility of the recipient was
Certainly all these were placebo effects, but the performed by Sherman J. Silber (Silber and Rodriguez-
world had obviously waited for such quackery, Rigau 1980) between twin brothers.
because in no time the extracts of animal organs by In Vienna, Eugen Steinach (18611944) per-
the Brown-Squard method were sold all over the formed vasoligation for rejuvenation (Steinach
(western) world, and factories sprung forth in Europe 1920), and one of his followers, Serge Voronoff
as well as in America, for example next to Central (18661951) turned to xenotransplantation and used
Park in New York (Borell 1976). There must have monkey testes to be transplanted for rejuvenation
been a real craze for these products, and physicians (Voronoff 1920; 1923). He first offered his surgery
concerned about the image of the young field of in Paris, but after some scandals continued his ques-
endocrinology started worrying. The famous neuro- tionable operations in Algiers, where he was obvi-
surgeon Harvey W. Cushing (18691939), and the ously visited by patients from all over the world.
president of the Association of the Study of Internal Voronoff had followers in many countries who xeno-
Secretions, E. H. Rynearson even talked about endo- transplanted animal testes or pieces thereof to
criminology in the context of this organotherapy patients in need of rejuvenation, also in the USA,
(Hamblen 1950). This assessment of the medical where this type of treatment caused great interest
scene at the time is also reflected in contemporary among the laymen and the media (e.g. Gayton
cartoons and comic songs from the early twentieth 1922). As unrest among the medical profession con-
century. Eventually, this type of quackery stimulated tinued to grow, in 1927 the Royal Society of Medicine
science and decent pharmaceutical companies to (London) sent an international committee to Voron-
search for real hormones. off in Algiers, which concluded that Voronoffs claims
were all poppycock (Parkes 1965). This, and the suc-
cess of upcoming steroid biochemistry finally termin-
1.3.2 Testis transplantations ated this malpractice.
However, before science succeeded in that attempt However, in a transformed fashion it continued as
there was another sad approach to treat hypogona- cellular therapy by injecting suspensions of fresh cells
dism and bring about rejuvenation and treatment of sheep embryos including testis cells, also for reju-
for all sorts of disorders: the transplantation of venation and revitalization, well into the second half
testes. G. Frank Lydston (18581923) in Chicago of the twentieth century (Niehans 1952; 1960).
was one of the first to perform human testicular Meanwhile, science has progressed and, in the age
transplantation from donors after experimentation of cell biology, testicular transplantation continues
in animals (Lydston 1915; Schultheiss and Engel with the aim of inducing fertility, but now uses iso-
2003). V. D. Lespinase (1913) published his experience lated germ cells (Brinster and Zimmermann 1994;
with transplanting human testes to patients for reju- Schlatt et al. 2002), and fertility has indeed been
venation, and Leo Stanley (1920) reported 20 cases of restored by this method in gamma-irradiated cocks
transplantation of testes from executed prisoners to (Trefil et al. 2006). Whether this may become a
other inmates who reported signs of revitalization. method to treat male infertility, for example in

7
Chapter 1: History of testosterone

Klinefelter patients (Wistuba 2010), remains to be various drug companies cooperating with scientists in
seen, but at least it is pursued on a rational scientific order to replace the miscredited organotherapy and to
basis as far as our present knowledge goes. bring proper hormone substitution to patients (see
Table 1.1) (Simmer 1982).

1.4 Twentieth-century science


and medicine 1.4.2 Testosterone is born
In 1931 Butenandt isolated the androgenic steroid,
1.4.1 The rise of steroid biochemistry androsterone (androstan-3a-ol-17-one), from urine,
Bertholds unique discovery was superseded by for which he required 15 000 liters provided by young
organotherapy, but it was not permanently forgotten:
Table 1.1 Early isolation of reproductive steroids (for references
Moritz Nussbaum (18501915), professor of anatomy see the text)
in Bonn, repeated Bertholds experiments and con-
1928 Guy Frederick Marrian Crystalline
firmed the results in frogs (Nussbaum 1909), as did
Pregnanediol Dept. Physiology and material from
Eugen Steinach in rats (Steinach 1910). Finally, Biochemistry, pregnant
A. Pzard confirmed Bertholds original results in University College mare urine
cocks (Pzard 1911), and the search for the active London provided by
androgenic substance in the testes began. BRITISH
From observation of the cocks comb growing under DRUG
the influence of transplanted testes, Moore et al. (1929) HOUSES
established the standardized capon combs test, meas- 1929 Estrone Edward Albert Doisy
uring androgenic activity in square centimeters of comb Dept. Biological
surface. This first bioassay facilitated determination of Chemistry, St. Louis
androgenic activity in body products as well as in chem-
1929 Estrone Adolf Butenandt Raw extracts
ical solutions. S. Loewe and H. E. Voss (1930) used the Chemistry Laboratory, and crystals
biological effects of androgens on the accessory sex University of from
organs and developed the cytological regeneration Gottingen pregnant
test, which was based on regrowth of the seminal mare urine
vesicle epithelium under androgenic substances provided by
(LoeweVoss test). The then still hypothetical male SCHERING
hormone was called androkinin. 1929 Estrone Ernst Laqueur Benzene
Simultaneously, steroid biochemistry emerged and Pharmacotherapeutic extracts from
the great breakthroughs were the discovery of the ring Laboratory, pregnant
structure of steroids and bile acids at the National Amsterdam mare urine
Institute of Medical Research in London (Rosenheim provided by
and King 1932) and at the Bavarian Academy of Sci- ORGANON
ences in Munich (Wieland and Dane 1932). A heated 1931 Adolf Butenandt Extracted
discussion started about whether there were three or Androsterone Chemistry Laboratory, from 15 000
four rings in the steroid structure and, if four rings, University of liters of urine
whether the fourth had five or six carbon atoms. Under Gottingen provided by
the sponsorship of the Health Organization of the the Prussian
League of Nations (the predecessor of the World Police
Health Organization, WHO), famous chemists includ- Academy in
ing Edmund A. Doisy, Adolf Butenandt and Guy Mar- Berlin and
processed by
rian assembled at University College London and
SCHERING
reached the consensus that steroids had four rings,
and the fourth ring had five carbon atoms (Butenandt 1935 Ernst Laqueur Isolated from
1980). Shortly before, these eminent researchers, Testosterone Pharmacotherapeutic bull testes
including Ernest Laqueur, had isolated pregnanediol Laboratory, Provided by
Amsterdam ORGANON
and estrone from pregnant mare urine provided by

8
Chapter 1: History of testosterone

policemen from Berlin, which was then processed steroid with pure anabolic effects on muscles or bones
by Schering to obtain 15 mg of this first androgen to treat cachexia, osteoporosis or small stature, or pure
(Butenandt 1931). In 1935 Ernst Laqueur (18661947) erythropoietic effect for the treatment of anemia with-
and his group in Amsterdam extracted and isolated 10 out androgenization could not be found. Nevertheless,
mg testosterone (androsten-17a-ol-3-one) from 100 kg anabolic and similar steroids were clinically used, but
of bull testes, which they found more active than disappeared again in the wake of evidence-based medi-
androsterone, and named it testosterone (David cine (Kopera 1985). However, they continued their
et al. 1935). In the same year Butenandt and Hanisch existence for illegal use and abuse for doping in sports
(1935) as well as Ruzicka and Wettstein (1935) and bodybuilding (see Chapters 24 and 25). Regret-
published the chemical synthesis of testosterone. This tably, at that time the pharmaceutical industry neg-
marked the beginning of modern clinical pharmaco- lected the chance to develop testosterone preparations
logy and endocrinology of testosterone, and male better suited for the substitution of hypogonadal
reproductive physiology. patients than the existing testosterone esters. It
Soon after its synthesis testosterone became clin- remains to be seen whether the current search for
ically available, first in the form of pellets (Deansley selective androgen receptor modulators (SARMs) will
and Parkes 1937; Parkes 1965) and then as injectable take a more rewarding course than did anabolic ster-
esters, that is, testosterone propionate, with a short oids (see Chapter 21).
half-life. And, from the mid-1950s on, the longer- From the 1970s the newly developed testosterone
acting testosterone enanthate (Junkmann 1952; immunoassays (see Chapter 4) made serial testoster-
1957) appeared, which remained the major testoster- one determinations in blood possible and, when
one preparation for half a century. Also in 1935, applied to pharmacokinetic studies, it turned out that
17a-methyltestosterone was synthesized and its oral all available testosterone preparations resulted in
effectiveness was demonstrated (Ruzicka et al. 1935). unphysiologically high or low serum levels which were
However, due to its 17a-structure it turned out to be undesirable in substitution therapy. Clinicians assem-
liver toxic (Werner et al.1950; Nieschlag 1981), a fact bled at a workshop on androgen therapy sponsored by
which gave testosterone in general a bad name among the WHO, and US National Institutes of health (NIH)
physicians, as this toxicity was also suspected for and Food and Drug Administration (FDA) in 1990
testosterone without reason; eventually in the 1980s came to the conclusion: The consensus view was that
this androgen became obsolete for clinical use in the major goal of therapy is to replace testosterone
Europe. In the late 1970s the orally effective testoster- levels at as close to physiologic concentrations as is
one undecanoate, absorbed from the gut via the possible (World Health Organization 1992), and
lymph to avoid the first-pass effect in the liver, was demanded that new testosterone preparations better
added to the spectrum of testosterone preparations suited for clinical use be manufactured.
used clinically (Coert et al. 1975; Nieschlag et al. In the mid-1990s, transdermal testosterone
1975) (see also Chapter 15). patches applied to the scrotal skin became the first
In the 1950s and 1960s the pharmaceutical indus- transdermal testosterone preparation in clinical use
try became more interested in new androgens than in (Bals-Pratsch et al. 1986). They had been invented by
testosterone itself, and concentrated its androgen Virgil Place at ALZA in Palo Alto, a company special-
research on the chemical modification of steroid mol- izing in new forms of delivery of known drugs (Atkin-
ecules in order to disentangle the various effects of son et al. 1998). However, although clinical results
testosterone and produce predominantly erythropoie- with this preparation were excellent, and for the first
tic or anabolic steroids (Kopera 1985). In 1956 con- time physiological serum levels could be achieved
temporary textbooks on androgens had already under testosterone substitution, physicians were
described 256 androgenic steroids (Dorfman and reluctant to prescribe a medication to be applied to
Shipley 1956), and by 1976 the number had increased the scrotum and preferred a subsequently developed
to more than 1000 (Kochakian 1976). non-scrotal system (Meikle et al. 1992). This, how-
However, it proved impossible to produce andro- ever, caused unpleasant skin reactions as it required
gens with only one effect out of the spectrum of testos- an enhancer to drive testosterone through the skin.
terone activities; at best, one of these effects could be For this reason the advent of the first transdermal
emphasized, but the other effects remained. The testosterone gel was welcome. This gel became

9
Chapter 1: History of testosterone

available in 2000 for the treatment of male hypo- knowing anything about the androgen receptor or
gonadism, first in the USA and later also in other androgen receptor mutations (see Chapter 3).
countries (Wang et al. 2000). Finally in 2004 At the same time, Huggins also posted his
the intramuscular testosterone undecanoate prepar- warning about testosterone influencing prostate
ation entered the market and soon achieved great carcinoma (Huggins and Hodges 1941), which led
popularity as a real testosterone depot preparation to castration (euphemistically called orchidectomy)
(see Chapter 15). as the major treatment of prostate carcinoma,
Testosterone undecanoate had originally been and prevailed until quite recently when androgen
used in oral capsules (see above), but had been turned deprivation therapy (ADT) by gonadotropin-
into an injectable preparation by Chinese investiga- releasing hormone (GnRH) analogs or antiandrogens
tors using tea seed oil as a vehicle (Wang et al. 1991). was introduced instead or in addition to castration
When the author came across it at an exhibition (maximal androgen blockade = MAB). Huggins
accompanying an andrology symposium in Beijing statement, Cancer of the prostate is activated
in 1993, samples were brought to Germany, injected by androgen injections induced a general fear of
into monkeys and showed a surprisingly long half- testosterone, especially among urologists, that pre-
life. A long half-life was confirmed in volunteering vented testosterone treatment in many patients
hypogonadal men who all showed serum levels in the who might have needed it. Only recently it became
normal range (Behre et al. 1999). When finally a clear that neither endogenous serum testosterone
company was interested in this fascinating prepar- levels (Endogenous Hormones and Prostate Cancer
ation, it was Europeanized by using castor oil as Collaborative Group 2008) nor testosterone treat-
vehicle and was developed as Nebido for clinical use ment (Raynaud 2006) have an impact on prostate
(Nieschlag 2006). carcinogenesis, and now testosterone treatment
under careful supervision is even considered for
1.4.3 Early descriptions of syndromes patients after radical prostatectomy suffering from
testosterone deficiency (Morgenthaler 2007). How-
of hypogonadism ever, orchidectomy will continue in the foreseeable
It may not have been a coincidence, but rather a sign future to remain the major option for treatment of
of heightened interest in the hypogonadism clinics, prostate carcinoma (Damber and Aus 2008). Thus,
that at the same time as testosterone became avail- castration continues to be recommended for thera-
able for clinical use (i.e. when rational treatment peutic purposes as it had already been promulgated
became possible), the major syndromes of primary during Greco-Roman times and the Middle Ages for
and secondary hypogonadism were first described: the treatment of leprosy, epilepsy, gout, priapism,
that is, the Klinefelter syndrome (Klinefelter et al. excessive masturbation and insanity (Melicow
1942) and the Kallmann syndrome (Kallmann et al. 1977), reflecting the knowledge or rather the lack of
1944). Pasqualini and Bur (1950) described the first knowledge of the respective period. There remains
case of the fertile-eunuch syndrome, characterized by the hope that research will eventually result in more
all symptoms of lack of testosterone, but active humane and patient-friendly methods of treatment
spermatogenesis; Del Castillo et al. (1947) published as we have witnessed the transition from castration
the first five cases suffering from Sertoli-cell-only to ADT for treating sexual offenders (Gooren 2011).
syndrome. Also at that time the symptoms of the
aging male were first described systematically, but
1.5 Key messages
unfortunately wrongly termed as male climacteric
(Werner 1945), starting a controversial discussion  The importance of the testes for normal male
that continues until today (see Chapter 16). function and reproduction has been known since
antiquity.
E. C. Reifenstein (1947) first described a syndrome
 Correspondingly, castration has also been
with partial androgen insensitivity (PAIS) that practiced since antiquity to produce obedient
carries his name to date, and J. M. Morris (1953) slaves, harem custodians, civil servants
published the first cases of complete androgen and soldiers, as well as for punishment and
insensitivity (CAIS) as testicular feminization in revenge.
male pseudo-hermaphroditism of course without

10
Chapter 1: History of testosterone

 Prepubertal castration was exercised between  In 1935 testosterone was chemically identified
the sixteenth and nineteenth century to obtain and synthesized and since then has been
voluminous high-pitched voices for singing in available for clinical use and also for abuse.
operas and churches.  In order to avoid inactivation in the liver and to
 Following the description of the morphological prolong action, testosterone has been esterified,
structure of the testes, their endocrine function and the clinically most widely used esters
was established by A. A. Berthold in 1849. became testosterone propionate, enanthate,
 Organotherapy with testes extracts and (xeno-) cypionate and finally undecanoate.
transplantation of testes were big business in the  Transdermal testosterone patches and gels
first half of the twentieth century, based on represent the most recent developments in
placebo effects. testosterone preparations.

1.6 References nineteenth century. Bull Hist Med


50:309320
active androgen. Acta Endocrinol
79:789800
Atkinson LE, Chang YL, Snyder PJ
(1998) Long-term experience Brinster RL, Zimmermann JW (1994) Damber JE, Aus G (2008) Prostate
with testosterone replacement Spermatogenesis following male cancer. Lancet 371:
through scrotal skin. In: Nieschlag E, germ-cell transplantation. Proc Natl 17101721
Behre HM (eds) Testosterone: Action, Acad Sci USA 91:12891302 David K, Dingemanse E, Freud J,
Deficiency, Substitution, 2nd edn. Brown-Squard E (1889) The effects Laquer E (1935) ber
Springer, Heidelberg, pp 365388 produced on man by subcutaneous krystallinisches mnnliches
Babtschenko A (2007) Die Farbe des injections of a liquid obtained from Hormon aus Hoden (Testosteron),
Krieges. Rowohlt, Hamburg the testicles of animals. Lancet wirksamer als aus Harn oder aus
20:105107 Cholesterin bereitetes Androsteron.
Bals-Pratsch M, Knuth UA, Yoon YD, Hoppe-Seylers Z Physiol Chem
Nieschlag E (1986) Transdermal Bundesrztekammer (ed) (2011) 233:281282
testosterone substitution therapy Placebo in der Medizin. Deutscher
for male hypogonadism. Lancet Deansley R, Parkes AS (1937) Factors
rzteverlag, Cologne
ii:943946 influencing effectiveness of
Butenandt A (1931) ber die administered hormones. Proc R Soc
Barker Jorgensen CB (1971) John chemische Untersuchung des London 124:279298
Hunter, A. A. Berthold and the Sexualhormons. Z Angew Chem
de Graaf R (1668) Virorum Organis
origins of endocrinology. Acta Hist 44:905908
Scient Natural Medicinal 24:154 Generationi Inservientibus,
Butenandt A (1980) Die de Clysteribus et de Usa
Barth H (1857) Reisen und Entdeckungsgeschichte des Siphonis in Anatomia. Ex officina
Entdeckungen in Nord-und Central- Oestrons. Endokrinol Inform Hackiana, Leyden and Rotterdam
Afrika in den Jahren 1849 bis 1855. 4:160163
Justus Perthes, Gotha Del Castillo EB, Trabucco A, de la
Butenandt A, Hanisch G (1935) ber Balze FA (1947) Syndrome
Bartoli C (2009) Sacrificium: the School Testosteron. Umwandlung des produced by absence of the
for Castratos, CD and booklet. Dehydroandrosterons in germinal epithelium without
Decca Music Group, London Androstendiol und Testosteron; ein impairment of the Sertoli or
Behre HM, Abshagen K, Oettel M, Weg zur Darstellung des Leydig cells. J Clin Endocrinol Metab
Hubler D, Nieschlag E (1999) Testosterons aus Cholesterin. 7:493502
Intramuscular injection of Hoppe-Seylers Z Physiol Chem Dorfman RL, Shipley RA (1956)
testosterone undecanoate for the 237:8998 Androgens: Biochemistry, Physiology
treatment of male hypogonadism:
Christensen AK (1996) A history of and Clinical Significance. John
phase I studies. Europ J Endocrinol
studies on testicular Leydig cells: the Wiley & Sons,Inc., New York
140:414419
first century. In: Payne AH, Hardy Endogenous Hormones and Prostate
Berthold AA (1849) ber die M, Russel LD (eds) The Leydig Cell. Cancer Collaborative Group (2008)
Transplantation der Hoden. Cache River Press, Vienna, IL Endogenous sex hormones and
Arch Anat Physiol Wiss Med:4246
Coert A, Geelen J, de Visser J, van der prostate cancer: a collaborative
Borell M (1976) Brown-Squards Vies J (1975) The pharmacology analysis of 18 prospective
organotherapy and its appearance in and metabolism of testosterone studies. J Ntl Cancer Inst
America at the end of the undecanoate (TU), a new orally 100:170183

11
Chapter 1: History of testosterone

Flaig E (2009) Weltgeschichte der characterized by gynecomastia, Melicow MM (1983) Castrati singers
Sklaverei. Verlag CH Beck, Munich aspermatogenesis, without A- and the lost cords. Bull NY Acad
Frigeni-Careddu M (1996) Il Leydigism, and increased excretion Med 59:744764
Condottiero: Vita, Avventure e of follicle-stimulating hormone. Mitamura T (1992) Chinese Eunuchs:
Batagglie di Bartolomeo J Clin Endocrinol 2:615627 The Structure of Intimate Politics.
Colleoni. Sperling and Kupfer Kochakian CD (ed) (1976) Anabolic- Charles E. Tuttle, Rutland, VT
Editori, Milan Androgenic Steroids. Springer, Moore CR, Gallagher TF, Koch FC
Gaeto, M (1999) The bicentennial of a Heidelberg (1929) The effects of extracts of
forgotten giant: Lazzaro Spallanzani Kopera H (1985) The history of testis in correcting the castrated
(17291799). Internatl Microbiol anabolic steroids and a review of condition in the fowl and in the
2:273274 clinical experience with anabolic mammal. Endocrinology 13:367374
Gayton B (1922) The Gland Stealers. steroids. Acta Endocrinol Suppl Morgenthaler A (2007) Testosterone
Lippincott, Philadelphia 271:1118 deficiency and prostate cancer:
Gooren LJ (2011) Ethical and medical Kremer J (1979) The significance of emerging recognition of an
considerations of androgen Antoni van Leeuwenhoek for the important and troubling
deprivation treatment of male sex early development of andrology. relationship. Eur Urol 52:623625
offenders. J Clin Endocrinol Metab Andrologia II:243249 Morris JM (1953) The syndrome of
96:36283637 Lehrs (1832) ber die Darstellungen testicular feminization in male
Hamblen EC (1950) Endocrinology or der Helena in der Sage und den pseudohermaphrodites. Am J Obstet
endocriminology. Some abuses of Schriftwerken der Griechen. In: Gynecol 65:11921211
endocrine therapy. Southern Med J Historische und literarische Niehans P (1952) 20 years of cellular
43:506509 Abhandlungen der Kniglichen therapy. Med Klin 47:216
Deutschen Gesellschaft zu
Henderson J (2005) Ernest Starling and Niehans P (1960) Introduction to
Knigsberg, Second Collection.
hormones: an historical Cellular Therapy. Pageant, New York
Borntrger, Knigsberg, p 108
commentary. J Endocrinol 184:510
Lespinase VD (1913) Transplantation Nieschlag E (1965) Otto Deiters (1834
Hirschfeld M (1927) Die Behandlung
of the testicle. J Am Med Assoc 1863). Med Welt 23:222226
der Impotenz in der rztlichen
18:251253 Nieschlag E (1981) Ist die Anwendung
Praxis. Institut fr
Leydig F (1850) Zur Anatomie der von Methyltestosteron obsolet?
Sexualwissenschaft, Berlin
mnnlichen Geschlechtsorgane und Dtsch Med Wschr 106:11231125
Holzhaider H (2011) Verbotene Liebe.
Analdrsen der Sugethiere. Ztschr Nieschlag E (2006) Testosterone
Sddeutsche Zeitung March 31, p 10
Wiss Zool 2:157 treatment comes of age: new
Huggins C, Hodges CV (1941) Studies options for hypogonadal men. Clin
Loewe S, Voss HE (1930) Der Stand der
on prostatic cancer: I. The effect of
Erfassung des mnnlichen Endocrinol 65:275281
castration, of estrogen and of
Sexualhormons (Androkinins). Klin Nieschlag E, Mauss J, Coert A,
androgen injection on serum
Wschr 9:481487 Kicovic P (1975) Plasma androgen
phosphatases in metastatic
Lydston GF (1915) Sex gland levels in men after oral
carcinoma of the prostate. Cancer
implantation. NY Med J administration of testosterone or
Res 293297
51:601608 testosterone undecanoate. Acta
Jenkins JS (1998) The voice of the Endocrinol 79:366374
castrato. Lancet 351:18771881 Medvei VC (1993) The History of
Clinical Endocrinology. Parthenon Nieschlag E, Cppers EJ, Wickings EJ
Junkmann K (1952) ber protrahiert Publishing Group, New York (1977) Influence of sex, testicular
wirksame Androgene. Arch Path development and liver function on
Pharmacol 215:8592 Meikle AW, Mazer NA, Moellmer JF,
the bioavailability of oral
Stringham JD, Tolman KG, Sanders
Junkmann K (1957) Long-acting testosterone. Europ J Clin Invest
SW, Odell WD (1992) Enhanced
steroids in reproduction. Rec Progr 7:145147
transdermal delivery of testosterone
Horm Res 13:389419 across nonscrotal skin produces Nieschlag E, Nieschlag S, Behre HM
Kallmann FJ, Schoenfeld WA, Barrera physiological concentrations of (1993) Life expectancy and
SE (1944) The genetic aspects of testosterone and its metabolites in testosterone. Nature 366:215
primary eunuchoidism. Am J Ment hypogonadal men. J Clin Endocrinol Nussbaum M (1909) Hoden und
Def 158:203236 Metab 74:623628 Brunstorgane des braunen
Klinefelter HF, Reifenstein EC, Melicow MM (1977) Castration down Landfrosches (Rana fusca). Pflgers
Albright F (1942) Syndrome the ages. NY State J Med:804808 Arch Eur J Physiol 126:519577

12
Chapter 1: History of testosterone

Ortkemper H (1993) Engel wider Sertoli E (1865) Dell esistenza di Sveinsson H (1969) Lifid er
Willen: die Welt der Kastraten. particolori cellule ramificate nei dsamlegt (autobiography).
Henschel-Verlag, Berlin canalicoli seminiferi del testicolo Helgafell, Iceland
Parkes AS (1965) The rise of umano. Il Morgagni 7:3140 The Last Emperor (1987) Film directed
reproductive endocrinology, 1926 Setchell BP (1974) The contributions of by Bernardo Bertolucci. Columbia
1940. J Endocrinol 34:XXXXXii Reinier de Graaf to reproductive Pictures
Pasqualini RQ, Bur GE (1950) biology. Europ J Obstet Gynec Trefil P, Micakova A, Mucksova J,
Sindrome hipoandrogenico con Reprod Biol 4:113 Hejnar J, Poplstein M, Bakst MR,
gametogenesis conservada. Shumankov VI, Gotsiridze OA (1978) Kalina J, Brillard J-P (2006)
Classificacion de la insufficiencia Chirurgische Technik der Restoration of spermatogenesis and
testicular. Rev Asoc Med Argent Hodentransplantation. Vestn Khir male fertility by transplantation of
64:610 Im II Grek 121:7781 dispersed testicular cells in the
Pzard A (1911) Sur la dtermination Silber SJ, Rodriguez-Rigau LJ (1980) chicken. Biol Reprod 75:575581
des caractres sexuels secondaires Pregnancy after testicular Tuchel S (1998) Kastration im
chez les gallinacs. Cpt Rend Scienc transplant: importance of treating Mittelalter. Droste-Verlag,
153:1027 the couple. Fertil Steril 33:454455 Dsseldorf
Podlech A (1990) Abaelard und Simmer HH (1980) Arnold Adolph Van Eickels K (2004) Gendered violence:
Heloise. Die Theologie der Liebe. Berthold (18031861) Vorlufer castration and blinding as
Piper, Munich oder Begrnder der experimentellen punishment for treason in Normandy
Raynaud JP (2006) Prostate cancer risk Endokrinologie? Endokrinol Inform and Anglo-Norman England. Gender
in testosterone-treated men. 3:101111 & History 16:588602
J Steroid Biochem Mol Biol Simmer H (1982) Biosynthese der van Leeuwenhoek A (1948) Collected
102:261266 Steroidhormone. Endokrinol Inform Letters of Antoni van Leeuwenhoek
Reifenstein EC Jr (1947) Hereditary 2:8092 Vol III. Swets and Zeitlinger,
familial hypogonadism. Proc Am Amsterdam
Simmer H, Simmer I (1961) Arnold
Fed Clin Res 3:86 Adolph Berthold (18031861). Zur Virdis R (2005) Historical milestones
Rosenheim SO, King H (1932) The Erinnerung an den hundertsten in endocrinology. J Endocrinol
ring-system of steroids and bile Todestag des Begrnders der Invest 28:944948
acids. Nature 130:315 experimentellen Endokrinologie. Voronoff S (1920) Testicular Grafting
Dtsch Med Wschr 86:21862192 from Ape to Man. Brentanos Ltd,
Ruzicka L, Wettstein A (1935)
Synthetische Darstellung des Spallanzani L (1779) Fecundazione London
Testishormons, Testosteron artefiziale. In: Prodomo Della Nuova Voronoff S (1923) Innesti Testicolori.
(Androsten 3-on-17-ol). Helv Chim Encyclopedia Italiana, Siena Quintieri Editore, Milan
Acta 18:12641275 Stanley LL (1920) Experiences in Wagner R (1852) Mitteilung einer
Ruzicka L, Goldberg MW, Rosenberg testicle transplantation. Cal State einfachen Methode zu Versuchen
HR (1935) Herstellung des 17alpha- J Med 18:251253 ber die Vernderungen
Methyl-testosterons und anderer Stanley LL (1923) An analysis of one thierischer Gewebe in
Androsten und thousand testicular substance morphologischer und chemischer
Androstanderivate. implantations. Endocrinol Beziehung. Nachr Knigl Ges Wiss
Zusammenhnge zwischen 7:787794 Gttingen:97109
chemischer Konstitution und Wang C, Berman N, Longstreth JA,
mnnlicher Hormonwirkung. Helv Starling EH (1905) The chemical
Chuapoco B, Hull L, Steiner B,
Chim Acta 18:14871498 correlation of the functions of the
Faulkner S, Dudley RE, Swerdloff
body I. Lancet 2:339341
Schlatt S, Foppiani L, Rolf C, RS (2000) Pharmacokinetics of
Weinbauer GF, Nieschlag E (2002) Steinach E (1910) Geschlechtstrieb und transdermal testosterone gel in
Germ cell transplantation into x- echt sekundre Geschlechtsmerkmale hypogonadal men: application of gel
irradiated monkey testes. Hum als Folge der innersekretorischen at one site versus four sites: a
Reprod 17:5562 Funktion der Keimdrsen. Zbl general clinical research center
Physiol 24:551566 study. J Clin Endocrinol Metab
Schultheiss D, Engel RM (2003) G.
Steinach E (1920) Verjngung durch 85:964969
Frank Lydston (18581923)
revisited: therapy by testicular experimentelle Neubelebung der Wang L, Shi DC, Lu SY, Fang RY
implantation in the early twentieth alternden Puberttsdrse. Springer- (1991) The therapeutic effect of
century. World J Urol 21:356363 Verlag, Berlin domestically produced testosterone

13
Chapter 1: History of testosterone

undecanoate in Klinefelter Wieland HO, Dane E (1932) Wistuba J (2010) Animal models
syndrome. New Drug Mark Untersuchungen ber die for Klinefelters syndrome and
8:2832 Konstitution der Gallensuren. their relevance for the clinic.
Hoppe-Sylers Ztschr Physiol Chem Mol Hum Reprod
Werner AA (1945) The male
210:268281 16:375385
climacteric. J Am Med Assoc
127:705710 Wilson JD, Roehrborn C (1999) Long- World Health Organization, Nieschlag
term consequences of castration in E, Wang C, Handelsman DJ,
Werner SC, Hamger FM, Kritzler RA men: lessons from the Skoptzy and Swerdloff RS, Wu F, Einer-Jensen
(1950) Jaundice during the eunuchs of the Chinese and N, Waites G (1992) Guidelines for
methyltestosterone therapy. Am Ottoman courts. J Clin Endocrinol the Use of Androgens. WHO,
J Med 8:325331 Metab 84:43244331 Geneva

14
Chapter
Testosterone: biosynthesis, transport,

2 metabolism and (non-genomic) actions


C. Marc Luetjens and Gerhard F. Weinbauer

2.1 Introduction 15 2.4 Testosterone transport 21


2.2 Leydig cells 15 2.5 Testosterone metabolism 23
2.3 Testosterone biosynthesis 16 2.6 Mechanism of androgen action 24
2.3.1 General aspects 16 2.6.1 Genomic effects of androgens 24
2.3.2 Regulation of testosterone synthesis by 2.6.2 Non-genomic effects of androgens 27
luteinizing hormone 19 2.7 Key messages 28
2.3.3 Regulation of Leydig cell function by factors 2.8 References 29
other than luteinizing hormone 21

2.1 Introduction refers to human data. It provides a timely overview of this


Androgens are essential for the development and func- topic and focuses on Leydig cells, regulation of Leydig cell
tion of male reproductive organs, for example matur- function, steroidogenesis, transport and metabolism of
ation of secondary sexual characteristics, libido and testosterone and genomic/non-genomic androgen
stimulation of spermatogenesis. Beyond that, andro- actions. For more detailed and extensive descriptions
gens influence many somatic organ functions, which on the various topics, the reader may also find the book
are covered in various chapters in this volume. In fact, The Leydig Cell in Health and Disease edited by Payne and
a large number of organs express androgen receptors Hardy (2007) useful.
(Dankbar et al. 1995). Physiological effects of andro-
gens depend on different factors such as number of
androgen molecules, distribution of androgens and 2.2 Leydig cells
their metabolites inside the cell, interaction with the The Leydig cells are located in the testicular interstitium.
receptors, polyglutamine number of the amino-acid These cells are the main source of testosterone, and
sequence in the androgen receptor and receptor acti- produce approximately 95% of circulating testosterone
vation (Palazzolo et al. 2008). In order to achieve suffi- in men. Approximately 1020% of the interstitial area is
cient exposure to androgens in target tissues, their occupied by Leydig cells, and the human testes contain
peripheral and local levels must be well balanced and approximately 200  106 Leydig cells. Daily testoster-
the transport mechanisms must be in place. Obviously, one production from the testis is estimated to be around
production and clearance/excretion rates must be in 67 mg. Since most of the peripheral testosterone is
balance as well. The action of androgens in target cells derived locally from the testicular Leydig cells, it is not
depends on the amount of steroid which can penetrate surprising that testicular testosterone concentrations
into the cells, the extent of metabolic conversions within exceed those of sex hormone-binding globulin
the cells, the interactions with the receptor proteins and, (SHBG)/androgen binding protein (ABP) by about
finally, upon the action of the androgen receptors at the 200-fold (Jarow et al. 2001), indicating a substantial
genomic level. Unless mentioned specifically, this chapter surplus of (free) testosterone in the testis. Relative to

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

15
Chapter 2: Biosynthesis, transport, metabolism and actions

concentrations in peripheral blood, testicular testoster- although the contribution of cells in the nervous
one concentrations are indeed approximately 80-fold system to circulating hormones is very small, local
higher (Coviello et al. 2005). The Leydig cell is the only production of steroids can be physiologically very
cell expressing all enzymes essential for converting important, especially when transport and clearance
cholesterol into testosterone (Payne 2007). In the Ley- are low (Rommerts 2004).
dig cell, testosterone can also be further metabolized
into dihydrotestosterone (DHT) or estradiol.
From the developmental, morphological and 2.3 Testosterone biosynthesis
functional viewpoint, different types of Leydig cells
can be distinguished: stem Leydig cells as founder 2.3.1 General aspects
cells, progenitor Leydig cells as committed stem cells, Since Leydig cells cannot store androgens, de novo
fetal Leydig cells as terminally differentiated cells in biosynthesis takes place continuously. The starting
the fetus, and adult Leydig cells as the terminally point for androgen synthesis is cholesterol, a funda-
differentiated Leydig cell (Ge and Hardy 2007). Fetal mental metabolic substance, with the typical steroid
Leydig cells become neonatal Leydig cells at birth ring conformation energetically compatible with the
and degenerate thereafter or regress into immature transformation into androgens (Fig. 2.1). Unlike most
Leydig cells (Prince 2007). Fetal Leydig cells produce cells that use cholesterol primarily for membrane
testosterone. Immature Leydig cells that mainly synthesis, Leydig cells have additional requirements
produce androstane-3a,17b-diol instead of testoster- for cholesterol, because it is the essential precursor
one have also been described. The proliferation rate for all steroid hormones. Luteinizing hormone, as
of the Leydig cells in the adult testis is rather low the central regulatory factor, controls both steroido-
and is influenced by luteinizing hormone (LH). The genesis and Leydig cell cholesterol homeostasis in
ontogeny of Leydig cells is not entirely clear, and vivo. Cholesterol can either be incorporated by the
mesonephros, neural crest and celomic sources have cell through receptor-mediated endocytosis from
been discussed. In the adult testis, Leydig cells develop low-density lipoproteins (LDLs), or can be synthe-
from perivascular and peritubular, mesenchymal-like sized de novo within the Leydig cell starting from
cells, and the differentiation of these cells into Leydig acetyl-coenzyme A. In addition, testosterone signaling
cells is induced by LH but also by growth factors and regulates lipid homeostasis in Leydig cells, and also
differentiation factors derived from Sertoli cells. Both affects the synthesis of steroids and modulates the
synthesis and secretion are under regulation of pituit- expression of genes involved in de novo cholesterol
ary LH and local factors (Lei et al. 2001; Sriraman synthesis (Eacker et al. 2008). Cholesterol is stored in
et al. 2005). In general terms, in most mammalian cytoplasmic lipid droplets. The number of lipid drop-
species, the testis goes through a period of independent lets is inversely related to the rate of androgen synthe-
function before the fetal hypothalamic-pituitary- sis, meaning that a high synthesis rate is accompanied
gonadal axis develops at around mid-gestation (see by a low content of lipid droplets and vice versa.
OShaughnessy and Fowler 2011 for review). Later, the Androgen synthesis requires the conversion of
fetal testes appear to become pituitary hormone depend- cholesterol to testosterone. This transformation is
ent, concurrent with declining Leydig cell function, but achieved via five different enzymatic steps, at the
increasing Sertoli cell numbers. Rodents and primates end of which the side chain of cholesterol is
appear to represent special cases insofar as rodents are shortened through oxidation from 27C to 19C. The
born well before the pituitary-dependent phase of fetal steroidal A-ring assumes a keto configuration at
development; whilst in primates the testis is dependent position 3. The starting point for the transformation
upon placental gonadotropin released during the pituit- of cholesterol into testosterone is the shortening of
ary-independent phase of development. the side chain through C22 and C20 hydroxylases,
Aside from Leydig cells, the remaining testoster- followed by cleavage of the bond between C20 and
one/androgen production with biological significance C22, leading to production of pregnenolone
is derived from adrenal steroidogenesis. The produc- (Fig. 2.2). The steps following pregnenolone forma-
tion of steroids is not necessarily limited to endocrine tion occur in the endoplasmic reticulum, either
glands, but very small amounts, mainly pregnane through the D4 or through the D5 pathway. The
derivatives, can also be produced in brain cells, and designation D4 or D5 refers to the localization of the

16
Chapter 2: Biosynthesis, transport, metabolism and actions

CH3 CH2 CH2 CH3


Cholesterol
CH CH2 CH
CH3
CH3
CH3

P450scc
HO

CH3 CH3
17-hydroxy- Dehydroepiandrosterone
Pregnenolone C O C O
pregnenolone O
P450c17
OH
P450c17

HO 3-HSD 3-HSD HO 3-HSD


HO

CH3 CH3
C O 17-hydroxy- C O
Progesterone Androstenedione O
progesterone
OH

CYP21A2 CYP21A2
O O O

CYP19 17-HSD

O OH
Estrone Testosterone

CYP19
HO O

17-HSD
OH

Estradiol

HO
Fig. 2.1 Steroidogenic pathways in the human testis.

double bond in the steroid. The D5 pathway is pre- stage, the intermediate androstene-3,17-dione is pro-
dominant over the D4 in human steroid synthesis. duced, which, through further reduction at position
Along the D4 pathway, pregnenolone is dehydrated C17, is then transformed into testosterone. In the D5
to progesterone, a key biological substance. The D4 synthesis pathway, testosterone synthesis occurs
pathway proceeds to the intermediate 17a-hydroxy- through the intermediates 17-hydroxypregnenolone
progesterone. If the side chain is removed at this and dehydroepiandrosterone (DHEA).

17
Chapter 2: Biosynthesis, transport, metabolism and actions

Fig. 2.2 Cholesterol uptake, transport to


LDL receptor the inner mitochondrial membrane and
mitochondrial side-chain cleavage system
yielding pregnenolone in mitochondria,
and subsequent metabolism of
Endoplasmic pregnenolone in endoplasmic reticulum.
reticulum Normally human steroidic cells take up
circulating low-density lipoproteins (LDL)
through receptor-mediated endocytosis,
directing the cholesterol to endosomes.
Cholesterol may also be stored in the
Endosome endoplasmic reticulum or it may be
esterified by acyl-CoA cholesterol
transferase and stored in lipid droplets as
cholesterol esters. Steroidogenic acute
regulatory protein (StAR) plays a key role
during steroidogenesis. StAR is localized
Cholesterol to the inner mitochondrial membrane
and governs cholesterol transport. StAR is
Transferase
responsible for the rapid movement of
cholesterol from the outer mitochondrial
membrane to the inner mitochondrial
Lipase
membrane, side chain-cleaved and
StarD4 converted by P450scc to pregnenolone.
Pregnenolone is biologically inactive and
StAR is processed further in the endoplasmic
Lipid
Adenodoxin
reticulum. Depending on activity and
reductase availability of other enzyme systems,
Adenodoxin
other steroids can then be produced
e further, or different pathways can be
e e employed for synthesis of the same
P450scc
Progesterone steroid hormone.
NADPH
Pregnenolone
3-HSD

Mitochondria P450
reductase
P450c17
3OHD/ 17-hydroxylase/
5-4 isomerase lyase

17OHD
Endoplasmic
reticulum
Testosterone

Cholesterol is the starting point for biosynthesis of regulatory protein mRNA expression is triggered by
steroids, oxysterols and bile acids. After cholesterol, endocrine stimuli and is rapidly and widely distrib-
an insoluble molecule is synthesized de novo or taken uted among steroidogenic tissues including adrenals
up via the LDL receptor or into the cell. Cholesterol and corpora lutea. Steroidogenic acute regulatory
available for steroidogenesis is stored in an ester form protein moves cholesterol from the outer to the inner
in lipid droplets, which are hydrolyzed by LH acti- mitochondrial membrane, but acts exclusively on the
vation of cholesterol ester hydrolase. Cholesterol outer membrane. The precise mechanism by which
needs to be transported intracellularly to mitochon- StARs action stimulates the influx of cholesterol
dria for incorporation into the mitochondrial cristae. remains unclear, but when StAR connects to choles-
The discovery of the steroidogenic acute regulatory terol it performs a conformational change that opens
protein (StAR) and associated proteins containing a cholesterol-binding pocket (Miller 2007). Following
StAR-related lipid transfer domains have helped phosphorylation, StAR interacts with voltage-dependent
substantially to unravel and understand this limiting anion channel 1 (VDAC1) on the outer membrane,
step of testosterone synthesis. Steroidogenic acute which processes the phospho-StAR into a smaller

18
Chapter 2: Biosynthesis, transport, metabolism and actions

intermediate. If VDAC1 is lacking, phospho-StAR is LH


degraded by cysteine proteases, preventing the mito-

chondrial membrane transport (Bose et al. 2008). Other
A number of StAR homologs have also been identi- Plasma membrane
G-proteins Effector receptors
fied with suspected roles in lipid binding and transfer
systems
activities. The physiological importance of StAR is LH receptor
highlighted by the phenotype of patients with an
inactivating mutation of the StAR gene. Being unable
to produce the necessary amounts of steroids, these
patients suffer from life-threatening congenital cAMP Ca2+/Calmodulin Diacylglycerol
adrenal hyperplasia (CAH) (King et al. 2011).
At the inner mitochondrial membrane site,
Protein kinases
cytochrome P450ssc (ssc = side-chain cleavage) cata-
lyzes the conversion of cholesterol into pregnenolone. Protein synthesis
The enzyme cytochrome P450ssc is responsible for
the different enzymatic reactions leading to the pro-
Growth Secreted
duction of pregnenolone. Like other steroid synthetic differentiation proteins
Steroidogenesis
enzymes, pregnenolone belongs to the group of mono- Fig. 2.3 Luteinizing hormone signaling in Leydig cells. Luteinizing
oxygenases, containing a prosthetic hemogroup in hormone binds to the LH receptor, a G-coupled membrane
hemoglobin, and localized on the internal membrane receptor. A great deal of information about the LH/LH receptor
interactions has been learned from studies dealing with receptor
of mitochondria. This reaction consists of three con- mutations. Since various effector systems are in place, it has been
secutive mono-oxygenations requiring two electrons assumed that factors other than LH can influence Leydig cell
to activate molecular oxygen: a 22-hydroxylation, steroidogenesis. Based upon Rommerts (2004).
20-hydroxylation and the cleavage of the C20C22
bond, yielding pregnenolone and isocaproic aldehyde. of testosterone (Figs. 2.3 and 2.4). Luteinizing
Pregnenolone diffuses across the mitochondrial mem- hormone is the most important hormone for regula-
branes and is transformed into testosterone through tion of Leydig cell number and functions (Rommerts
the enzyme cytochrome P450c17, also belonging to the 2004).
group of mono-oxygenases and located in the endo- Luteinizing hormone is also the key hormone
plasmic reticulum. Overall, however, the enzymatic stimulating Leydig cell testosterone synthesis and
system is not capable of transforming every molecule release via specific receptors. This concept received
of pregnenolone into testosterone, thus rendering sev- compelling support from recent clinical and molecu-
eral metabolic intermediates. lar observations. Single mismatch mutations in the
transmembrane region were associated with a consti-
tutive activation of the LH receptor in the absence
2.3.2 Regulation of testosterone synthesis of its ligand, and patients bearing this LH receptor
by luteinizing hormone defect showed familial precocious puberty, that is, the
Luteinizing hormone (LH) is a dimeric glycoprotein premature maturation of the gonad (Kremer et al.
containing a glycosylated polypeptide a-subunit 1993; Canto et al. 2002). There seems to be no func-
and b-subunit that are covalently linked. Luteinizing tional necessity for LH in the male until the onset
hormone binds to specific receptors in the testis of puberty. The first wave of spermatogenesis in
which are located on Leydig cells, and has a molecu- fetal mice occurs without LH or relies on maternal
lar weight of 93 kDa. The LH receptor like the human chorionic gonadotropin (hCG) in male fetuses
follicle-stimulating hormone (FSH) and GnRH (Huhtaniemi 2000). Luteinizing hormone receptor
receptor belongs to the family of G-coupled recep- knock-out (LHRKO) mice have normal testes at birth,
tors. Common to these receptors are seven trans- and the number of Leydig cells is identical to that of
membrane domains that are crucial for the normal control animals (Zhang et al. 2001; Rao and Lei 2002;
functioning of the gonadotropin receptors. The Lei et al. 2004). Human chorionic gonadotropin is
main action of LH is to bind to its receptor in the also a very potent stimulator of Leydig cell testoster-
Leydig cells and to induce the synthesis and release one production. Testosterone is already produced by

19
Chapter 2: Biosynthesis, transport, metabolism and actions

Fig. 2.4 Endocrine and local regulation


Hypothalamus GnRH of Leydig cell steroidogenesis by LH and
Brain
locally produced factors. Luteinizing
hormone is the key regulator of Leydig
Kisspeptin cell function, and testosterone is the key
product of the Leydig cell. A sensitive and
rapid negative-feedback loop exists
Pituitary between the Leydig cells and the brain
that is mediated via testosterone (and its
metabolites DHT and estradiol). Another
key product of the Leydig cells is insulin-
like factor 3 (INSL3), playing an important
LH role in testicular development and
descent. A variety of factors, for instance a
FSH plethora of growth factors, is derived
from the seminiferous tubules and
Growth modulates Leydig cell steroidogenesis
factors these effects can either be stimulatory or
inhibitory.
Leydig cells

Sertoli cells

INSL3

Gonad

Testes

Tubule
Testosterone

the fetal testis during the 10th week of gestation, stimulating testosterone production, because in
under the stimulation of fetal LH and maternal studies administering exogenous LH, pulsatile tes-
hCG. The role of maternal hCG in this crucial phase tosterone production seems to increase before LH
of gonadal development is suggested by the fact that a reaches the Leydig cells through the interstitial space
mutation of the LH b chain leading to a biologically (Setchell et al. 2002; Setchell 2004). Also, although
inactive gonadotropin is associated with normal LH receptors were believed to be expressed in the
sexual differentiation. Conversely, inactivating muta- gonads only, a number of reports have documented
tions of the LH receptor produce a clinical syndrome the presence of LH receptors in a variety of organs
resembling complete androgen insensitivity, with a and tissues. The rat prostate expresses a functional
phenotype of female external genitalia (Themmen gonadal-sized receptor capable of binding iodinated
et al. 1998). hCG and stimulating a cAMP response in a LH dose-
It has been shown that LH receptors are not only dependent manner. The main expression was seen in
confined to the Leydig cells within the male gonad. the ventral lobe, which is highly dependent on
The presence of LH/hCG receptors has been demon- androgens. Tao et al. (1997) have also demonstrated
strated in endothelial cells of the testicular micro- that the human prostate expresses luteinizing hor-
vasculature (Ghinea and Milgrom 2001). The mone receptors. These findings raise the possibility
relevance of vascular LH/hCG receptors for testicu- that, in the male, LH exerts extragonadal actions and
lar physiology is currently unknown (Haider 2004). influences prostatic functions which still remain
Luteinizing hormone may not be the only factor undefined.

20
Chapter 2: Biosynthesis, transport, metabolism and actions

2.3.3 Regulation of Leydig cell function lending further support to the importance of local
interactions between Sertoli cells, Leydig cells and
by factors other than luteinizing hormone peritubular cells in connection with the actions of
Insulin-like factor 3 (INSL3) is a relaxin-like androgens and gonadotropins.
proteohormone produced by Leydig cells (Foresta
et al. 2004) and signals through a G-coupled receptor
(LGR8) that is expressed in Leydig cells and meiotic/ 2.4 Testosterone transport
postmeiotic testicular human germ cells but not in During transport in plasma, testosterone is mainly
peritubular and Sertoli cells (Anand-Ivell et al. 2006). bound to albumin or to SHBG which is produced by
Compelling evidence is available to indicate that hepatocytes. Androgen-binding protein, with similar
INSL3 is a marker of Leydig cell differentiation steroid-binding characteristics when compared to
and of entry into male puberty (Ferlin et al. 2006; SHBG, is produced by Sertoli cells in the testis, and
Wikstrm et al. 2006). Levels of INSL3 are influenced is a b-globulin consisting of different protein sub-
by hCG/LH, but this effect appears uncoupled from units. In rats, SHBG is expressed in Sertoli cells,
the steroidogenic effects of LH on testosterone syn- secreted preferentially into the seminiferous tubules,
thesis (Bay et al. 2006). Since receptors for INSL3 and migrates into the caput epididymidis where
are present on advanced germ cells, it is tempting to it is internalized by epithelial cells and modulates
assume a local role for INSL3 during spermatogen- androgen-dependent sperm maturation. Testicular
esis. While the role for spermatogenesis may be SHBG isoforms are found in sperm and released from
unclear, it appears that INSL3 provides a marker for sperm during the capacitation reaction. Plasma SHBG
Leydig cell capacity (Ivell and Anand-Ivell 2011). has about 95 kDa molecular weight, 30% of which is
Growth factors, at least by inference from in-vitro represented by carbohydrates, and possesses one
studies, appear to influence Leydig cell function androgen binding site per molecule. Human testicular
(Chandrashekar and Bartke 2007). Thus, studies have SHBG transcripts are expressed in germ cells and
suggested that the local function of inhibin and activin contain an alternative exon 1 sequence, appearing to
could be a modulation of steroidogenic activity encode an SHBG isoform that is 45 kDa smaller than
in Leydig cells. Activins inhibit or stimulate Leydig plasma SHBG. The testosterone binding capacity is
cell steroidogenesis in a species-dependent manner. also much lower compared to the plasma SHBG
Generally, insulin-like growth factor 1 (IGF-1) and (Selva et al. 2005). In normal men, only 2% of total
transforming growth factor alpha (TGF-a) exert a testosterone circulates freely in blood, while 44% is
stimulatory activity in the testis, while TGF-b acts as bound to SHBG and 54% to albumin. The binding
an inhibitor. In the rat, the development of Leydig affinity of testosterone to albumin is about 100 times
cells is sustained by an interplay between TGF-a and lower compared to SHBG. However, since albumin
TGF-b, and LH activity is modulated by IGF-1. In concentration is far higher than that of SHBG, the
the human Leydig cell the steroidogenic activity is also binding capacity of both proteins for testosterone
stimulated by epidermal growth factor (EGF). Thyroid is approximately the same. The ratio of testosterone
hormone accelerates the differentiation of Leydig cells bound to SHBG over free SHBG is proportional
(Ariyaratne et al. 2000) and also stimulates the StAR to SHBG concentration. A direct measurement of
expression and steroid production in fully developed free testosterone is impractical in routine practice,
cells (Manna et al. 1999). Glucocorticoids inhibit ster- so that several equations are used to estimate the free
oidogenic enzymes and induce apoptosis in rat Leydig testosterone concentration in serum (see Chapter 4).
cells (Gao et al. 2002). The ultimate effects of gluco- Apparently, dissociation of testosterone from
corticoids in vivo depend very much on local metab- binding proteins takes place predominantly in capil-
olism within the target cells. In this connection, laries. The interaction of binding proteins with the
the activity of 11b-hydroxysteroid dehydrogenase as endothelial glycocalyx leads to a structural modifica-
a local amplifier of glucocorticoid action is also of tion of the hormonal binding site and thereby to a
importance (Seckl and Walker 2001). Interestingly, change in affinity. As a result testosterone is set
recombinant FSH stimulates testosterone production free and can diffuse freely into the target cell, or
in men (Levalle et al. 1998) and in patients with binds together with SHBG to megalin (Fig. 2.5), a cell
selective FSH deficiency (Lofrano-Porto et al. 2008), importer protein (Hammes et al. 2005). Megalin is

21
Chapter 2: Biosynthesis, transport, metabolism and actions

(A) (B) Fig. 2.5 Mechanisms by which steroidal


LDL hormones can enter cells. (A) The steroid
Steroid carrier hormone diffuses freely across the cell
LDL receptor membrane, binds to an intracellular
receptor and enters the nucleus to
regulate gene expression. (B) Receptor-
mediated endocytosis of steroid
containing lipophilic molecules. The LDL
Steroid binds its receptor and is taken up,
receptor Steroid
Endosome degraded in lysosomes and the steroid
cholesterol can enter different metabolic
pathways. (C) Receptor-mediated
endocytosis of steroids. The entire
hormone carrier is endocytotically bound
after binding to a carrier protein.
Nucleus Following the intracellular degradation of
Cholesterol the carrier, the ligand hormone is
Intracellular Intracellular
Lysosome released into the free cytoplasm.
(D) Transport-mediated uptake of
molecules through the membrane. The
Extracellular steroid carrier is recognized by a
membrane receptor and the ligand is
(C) Steroid carrier (D) Steroid carrier transported into the cell. See plate section
for color version.
Megalin Steroid channel

Steroid
receptor

Endosome Nucleus Steroid

Nucleus
Intracellular Intracellular

expressed in sex-steroid target tissues and is a member of hormone-binding globulin concentration in serum
the LDL receptor superfamily of endocytotic proteins. In is under hormonal regulation and primarily regu-
the serum, 9899.5% of the sex steroids are protein lated through opposing actions of sex steroids on
bound, and endocytosis is quantitatively more relevant hepatocytes: estrogen stimulates and androgen inhibits
for tissue delivery of biologically active steroid hormones SHBG production. Other hormones such as thyroid
than free diffusion. To date several different ways have hormones are also potent stimulators of SHBG pro-
been described by which steroids can enter the target duction. Sex hormone-binding globulin concentration
cells, and which of these are the most relevant pathways in men is about one-third to one-half of the concen-
to take up the various steroid hormones is still being tration found in women. In normal, healthy men with
debated. an intact hypothalamic-pituitary-testicular axis, an
Sex hormone-binding globulin binds not only tes- increase in plasma concentrations of SHBG leads to
tosterone but also estradiol. The type of binding an acute decrease of free testosterone and simultan-
is influenced by the different SHBG isoforms, but eous stimulation of testosterone synthesis, persisting
generally testosterone binds threefold higher than until achievement of normal concentrations.
estradiol to SHBG. For example, it could be demon- Testosterone concentrations in the testicular
strated that post-translational changes in the carbohy- lymphatic circulation and in the venous blood are
drate structure of SHBG can lead to different binding very similar, but there are essential differences in the
affinity of the protein to testosterone or estradiol. Sex flow rate and velocity of both systems. Therefore,

22
Chapter 2: Biosynthesis, transport, metabolism and actions

transport of testosterone in the general blood circula- Transport


Biosynthesis Metabolism
tion occurs mainly through the spermatic vein. (plasma)
Androgens diffuse into interstitial fluid and then
enter testicular capillaries or enter capillaries directly Testis Protein bound Inactive
(95%) (98%) metabolites
from Leydig cells that are in direct contact with the
testicular microvasculature. The mechanism for tes-
Liver
tosterone transport from the Leydig cell into the
blood or lymph is not completely known. Probably
Testosterone Free testosterone
lipophilic steroids distributed within cells or small cell (2%) Testosterone
(6 mg/day)
groups are released through passive diffusion. On the
other hand, mouse studies have raised the possibility Peripheral
(prostate)
of an active testosterone transport being important
for spermatogenesis (Takamiya et al. 1998), showing Protein bound
Peripheral
that gangliosides-associated testosterone transport (99%) Dihydrotestosterone
(5%)
appeared necessary for complete spermatogenesis.
Steroids such as pregnenolone, progesterone and
testosterone not only rapidly pass the Leydig cell
membranes, but they can also equilibrate rapidly Free testosterone
between different testicular compartments, and the (1%)
testicular secretion pattern is most likely determined Fig. 2.6 Quantitative aspects of production, transport and
by amounts that are produced inside the tissue, metabolism of biologically active androgens. The majority of
the permeability characteristics of the membranes testosterone is derived from the testis. During peripheral transport,
most of the testosterone is bound to transport proteins. In
and the binding proteins in various testicular fluids peripheral tissues, testosterone is either metabolized or converted
(Rommerts 2004). As the blood flow is much higher into other active steroids such as DHT or estradiol. Based upon
than the flow of interstitial fluid, most of the uncon- Rommerts (2004).
jugated steroids diffuse from the interstitial space to
the blood and leave the testis via venous blood. (Luetjens et al. 2006; Modi et al. 2007). Using
Estradiol is produced by Leydig cells, but the amount a derivative of progesterone, norethisterone enanthate,
is small, with about 20% of peripheral aromatization no direct effects on testicular/epididymidal function
(Rommerts 2004). were found (Junaidi et al. 2005). Through 5a-reduction,
testosterone gives rise to DHT which is three- to
sixfold more potent than testosterone. The half-life
2.5 Testosterone metabolism of testosterone in plasma is only about 12 minutes.
Testosterone is the main secretory product of the Estrogens influence testosterone effects by acting
testis, along with 5a-dihydrotestosterone (DHT), either synergistically or antagonistically. Moreover,
androsterone, androstenedione, 17-hydroxyprogester- estrogens have other specific effects which were ori-
one, progesterone and pregnenolone (Fig. 2.6). The ginally described to be typical of testosterone. For
transformation of testosterone into DHT takes place example, it has been found that inactivating muta-
principally in the target organs, for example prostate. tions of the estrogen receptor or aromatase activity,
Androstenedione is important as a precursor for the preventing estrogen action on the bones, results in
production of extratesticular estrogens. Biologically continuous linear growth and lack of epiphyseal
active estradiol can be produced as a result of extra- closure.
testicular aromatization of androstenedione to estrone Low levels of bioavailable estrogen and testosterone
that is subsequently reduced to estradiol in peripheral are strongly associated with high bone turnover, low
tissues. Only a very small portion of the testosterone bone mineral density and high risk of osteoporotic
produced is stored in the testis, and the androgen is fractures. In aromatase-knock-out and estrogen recep-
mainly secreted in blood. The role of androsterone, tor-knock-out male mice, an association between
progesterone and 17-hydroxyprogesterone in the testis impaired glucose tolerance with insulin resistance and
is unknown, but progesterone receptors have been lack of estrogens with elevated testosterone concentra-
found in some peritubular cells and on spermatozoa tions has been found (Takeda et al. 2003). It also seems

23
Chapter 2: Biosynthesis, transport, metabolism and actions

that aromatase deficiency in men is associated and DHT. Changes in the property of type 2
with the occurrence of insulin resistance and diabetes 5a-reductase due to mutation can result in complete
mellitus type 2 during high-dose testosterone treat- androgen insensitivity syndrome (CAIS) or partial
ment (Maffei et al. 2004) (see also Chapter 11). The androgen insensitivity syndrome (PAIS) (see also
impairment of the estrogen-to-testosterone ratio is Chapter 3). Dihydrotestosterone is eliminated by type
thought to be responsible for the development of 3a-HSD (hydroxysteroid dehydrogenase; aldo-keto
impaired glucose tolerance and insulin resistance in reductase (AKR)1C2), which reduces 5a-DHT to
aromatase-deficiency patients receiving a testosterone 3a-androstanediol. Studies in men have demonstrated
replacement therapy (TRT). An imbalance in the ratio that 3a-androstanediol can be converted back to
of estrogen to androgen tissue levels is also postulated 5a-DHT by AKR1C2 activity only, to stimulate
as a major cause in the development of gynecomastia. growth of the prostate (Penning et al. 2000). In
Furthermore, recent investigations have shown a local human tissues, five aldo-keto reductase isoforms exist
neuroprotective effect of newly aromatized estradiol with varying reductase activity on the 3-, 17- and
on the brain. 20-ketosteroid position, with isoform AKR1C2 pre-
Reduction of testosterone to DHT occurs in the dominately converting 5a-DHT to 3a-diol. The inacti-
endoplasmic reticulum via the enzyme 5a-reductase vated metabolites are excreted in the urine. Some
located in the microsomes. Testosterone and DHT androgen metabolites are excreted in their free form;
both bind to the same intracellular androgen receptor others are glucuronated by the liver before excretion.
to regulate gene expression in the target tissue. Recently excreted metabolites have been used in
Although both hormones interact with the same screening assays to uncover doping with exogenous
androgen receptor, testosterone and DHT produce testosterone esters in high-performance athletics
distinct biological responses, and the molecular mech- (Saudan et al. 2006). The 17-glucuronidation of the
anisms that convey this differential activity are still DHT metabolite androstane-3a,17b-diol is correlated
under debate. Two isoforms of 5a-reductase have been with several metabolic risk factors in men. The ratio of
identified in humans. Being nicotinamide adenine 17G to DHT is associated with total fat mass, its distri-
dinucleotide phosphate (NADPH)-dependent enzymes, bution, intrahepatic fat, disturbed lipid profile, insulin
reduction of the double bond at the four to five position resistance and diabetes (Vandenput et al. 2007).
in C19 steroids as well as C21 steroids occurs. The gene
for 5a-reductase type 1 is located on chromosome 5,
encoding for a protein with 259 amino acids, while the 2.6 Mechanism of androgen action
gene for the 5a-reductase type 2 is on chromosome 2,
encoding for a slightly shorter protein with 254 amino 2.6.1 Genomic effects of androgens
acids. The two isoforms are very similar but show Testosterone dissociates from SHBG at the target organ
different biochemical properties. Type 1 enzyme works and diffuses into the cells. The conversion of testoster-
optimally at an alkaline pH, while the optimal pH for one into DHT is organ dependent. This occurs, for
type 2 is acidic. Also, the tissue distribution of the two example, in the prostate, where DHT is the main
forms is different. Type 1 5a-reductase has been local- biologically active androgen. Hydroxysteroid dehydro-
ized in the non-genital skin, liver, brain prostate, ovary genases regulate ligand access to the androgen receptor
and testis; while type 2 is mainly active in classical in the human prostate. The first step in androgen
androgen-dependent tissues, such as the epididymis, action is binding to the androgen receptor which
genital skin, seminal vesicle, testis and prostate, but also belongs to the family of steroid hormone receptors.
in liver, uterus, breast, hair follicles and placenta. At a Along with RoDH, HSDs are involved in the pre-
cellular level, DHT sustains differentiation and growth receptor regulation of androgen action. The androgen
and is particularly important for normal sexual devel- receptor and other nuclear receptors act as transcrip-
opment and virilization in men. It also affects muscle tional factors via a general mechanism in which they
mass and the deepening of the voice. Dihydrotestoster- bind to their cytosolic ligand, inducing conforma-
one transactivates the androgen receptor, leading to tional changes, loss of chaperones, dimerization and
prostate gene transcription and growth. nuclear translocation.
Overall, testosterone effects result from influences In the nucleus both (ligand and nuclear receptor)
of the hormone itself and of its metabolites estradiol bind to specific sequences of genomic DNA and

24
Chapter 2: Biosynthesis, transport, metabolism and actions

induce stimulation of RNA synthesis. Chromatin three short tandem repeats (STRs) CAG, coding for
remodeling such as modification of histones plays a polymorphic polyglutamine, TGG repeats coding for
role in gene transcription, and many nuclear receptor polyproline and GGC repeats for polyglycine. In
interacting coregulators perform significant roles in normal men, about 1729 glutamine repeats and
gene transcription. Histone demethylases are key 1317 glycine repeats are present. Alleles of small
players in the regulation of androgen-mediated tran- GGC size have been associated with esophageal
scription. In the absence of androgens, target genes cancer; while in patients with Kennedy disease, a
are turned off due to the presence of H3K9 methyla- disease with degenerating motoneurons, up to 72
tion of a lysine residue on histones H3 and H4. In the such glutamine repeats are present. Furthermore, in
presence of androgens, H3K9 methylation is removed, the androgen receptor, long CAG and GGC alleles are
which leads to the derepression of androgen target associated with decreased transactivation function
genes (Klose and Zhang 2007). For further detailed and have been associated with cancers in women. In
information please see the review by Wu and Zhang the androgen receptor, low-size CAG (<19 repeats)
(2009). Mineralocorticoid, glucocorticoid, thyroid and GGC (<15 repeats) alleles result in higher recep-
hormone, retinol, fatty acid metabolite, estrogen and tor activity, and have been associated with earlier
progesterone receptors also belong to this family. At age of onset, and a higher grade and more advanced
least 48 nuclear receptors have been identified in stage of prostate cancer at the time of diagnosis.
humans (Kishimoto et al. 2006). These receptors share The number of glutamine repeats of the androgen
substantial functions and are thought to have evolved receptor has been associated with azoospermia or
from a single ancestral gene (Bertrand et al. 2004). oligozoospermia, but no clear association was found
Orphan nuclear receptors have also been found for (Asatiani et al. 2003; see also Chapter 3).
which no ligand has yet been identified. Members of The androgen receptor gene encoding for two
this receptor family possess an N-terminal domain, a isoforms is located on the X chromosome and spans
DNA-binding domain (DBD), a hinge region and a about 90 kb and codes for a 2 757-base pair open
hormone-binding domain. reading frame within a 10.6-kb mRNA. The location
Nuclear receptors have several functional of the androgen receptor gene on the X chromosome
domains, a constitutionally activating function is preserved in evolutionarily distant animals, such as
domain, a highly conserved DNA-binding domain, a marsupials and monotremes, and may reflect a devel-
nuclear localization signal domain and a ligand- opmentally significant association of the androgen
binding domain (LBD). Steroid receptors show high receptor gene with other systemic genes. The coding
homology with the corresponding DNA-binding and sequence, containing the sequence of nucleotides
ligand-binding domains in the mineralocorticoid, translated into amino acids, consists of eight exons.
glucocorticoid and progesterone receptors. In con- The N-terminal domain, the transcriptional regula-
trast, at the N-terminal domain little similarity with tory region of the protein, is fully encoded by exon 1,
these receptors remains. The nuclear receptors are while DNA-binding domain is encoded by exon 2 and
subdivided into two subfamilies depending on their 3, and steroid-binding domain by exons 48.
ligand partners forming homodimers, such as the Similar to all the other steroid receptors, the
androgen receptor and other steroid receptors; DNA-binding domain of the androgen receptor con-
another subfamily forms heterodimers with only one tains two zinc fingers. This domain is about 70 amino
ligand, such as the thyroid hormone receptor. In most acids long and is localized between the N-terminal
nuclear receptors the binding in a pouch to their and the androgen-binding domain. In this part of
ligand at the C-terminal end, made of 12 a-helices, the sequence, eight cysteines are spatially arranged
leads to a shift of the most C-terminal a-helix and to such that four sulfur atoms keep one zinc atom in
an alteration of its activity. Inside the 12 helices, a place, giving rise to the typical structure of two over-
hydrophobic cave is induced to change its formation lapping helices. Exon 2 and 3 of the androgen recep-
and angle which the helix number 12 folds into. tor encode for the DNA-binding domain. The first
This angle is associated with transactivation status of zinc finger, encoded by exon 2, is important for the
the receptor. specific binding of the androgen receptor to the
An important characteristic of the N-terminal second and fifth nucleotide pairs in the first andro-
domain of the androgen receptor is the presence of gen-response element repeat GGTACA of the DNA.

25
Chapter 2: Biosynthesis, transport, metabolism and actions

The second zinc finger, encoded by exon 3, stabilizes factor importin-b. The importin-aimportin-b
DNA receptor binding by hydrophobic interactions androgen receptor complex then moves through
with the first finger, and contributes to specificity of nuclear-pore complexes and dissociates, mediated by
receptor DNA binding, leading to dimerization of two a guanosine triphosphatase (GTPase) protein, thus
receptor molecules. In immediate proximity to the releasing the androgen receptor inside the nucleus.
DNA-binding domain, a short amino-acid sequence The actual interaction with DNA occurs through
is responsible for the transport of the receptor into the two zinc fingers in the DNA-binding domain
the nucleus. of the androgen receptor. The androgen receptor
The androgen-binding domain of the androgen interacts with DNA in a homodimeric form consis-
receptor encompasses about 30% of the overall recep- ting of two identical hormone receptor complexes.
tor (the DNA-binding domain about 10%) and is These homodimers bind to DNA sequences known
responsible for the specific binding of androgens. as androgen-responsive elements, which contain typ-
This domain forms a lipophilic pocket based upon ical palindromic sequences, that is, DNA tracts with a
24 amino acids that enables the binding of testoster- nucleotide sequence independent of the reading direc-
one or other androgens. Experimental deletion of this tion. Before it can bind to the DNA, covalent modifi-
domain results in increased gene transcription in cations of the DNA trapping proteins, the histones,
vitro. This part of the receptor is necessary for indu- have to occur first. At the N-terminal ends of histones
cible and regulated gene transcription. Regulation of various covalent modifications can be carried out,
steroid hormone receptor action occurs, in part, by such as acetylation, phosphorylation, ubiquitination
post-translational modifications, such as phosphoryl- and methylation. Depending on the histone modifi-
ation. The androgen receptor is phosphorylated at a cations, genes will either be expressed or silenced, and
number of sites in response to agonist binding that the methylation of lysines or arginines has been linked
results in nuclear localization, but usually not in to either transcriptional activation or repression.
response to antagonists. It has also been shown that Lysine residues of the histone tails can be monomethy-
phosphorylation is regulated by steroid hormones or lated, dimethylated or trimethylated. The differentially
forskolin and phorbol esters at different sites. Dihy- methylated lysine residues serve as binding sites for
drotestosterone binds to the androgen receptor with various effector proteins which may be co-repressor
higher affinity than testosterone, mainly because tes- or coactivators.
tosterone more quickly dissociates from the receptor. Lysine-specific demethylase 1 (LSD1) has the func-
Other steroids such as androstenedione, estradiol and tion to silence genes if no nuclear receptor is present
progesterone bind to the androgen receptor with but, in the opposite case, to demethylate histones
much lower affinity than testosterone. Phosphoryl- if such a ligand-activated receptor is docking onto
ation in the hinge region required for full transcrip- the site. During this step, LSD1 forms a chromatin-
tional activity is involved in the signal transduction associated complex with the ligand-activated androgen
mediated by cyclic AMP and protein kinase C. receptor. Another coactivator is needed to start actual
Chaperones such as heat shock protein 90 (HSP gene expression of the LSD1 chromatin-associated,
90) are responsible for the maintenance of the recep- ligand-activated androgen receptor complex. This
tor in the inactive state and are released from the again is a demethylase (JHDM2A) which is only
complex. Without its hormone ligand the androgen associated with DNA in combination with the above-
receptor can bind to homologous or heterologous mentioned complex. Two different mechanisms involv-
ligands in the cytoplasm and exert inhibitory effects ing demethylases in the transcriptional regulation of
on cell death processes. The loss of this protein has the androgen receptor are needed to express androgen
uncovered functional domains of the receptor and is target genes (Metzger et al. 2006). The time sequence
necessary for nuclear transport. Nuclear transport is of events is not known. Up to now many methylation/
mediated by import receptors through a pathway that demethylation enzymes have been found which all may
employs two proteins, importin-a and importin-b. In be needed for fine tuning of gene expression. The
this nuclear-protein-import pathway, the positively transcription complex induces mRNA synthesis of
charged nuclear localization signal of the androgen androgen-dependent genes and, after mRNA transla-
receptor is recognized in the cytoplasm by importin-a tion, synthesis of new androgen-dependent proteins
that serves as an adaptor to the nuclear transport such as prostate-specific antigen (PSA).

26
Chapter 2: Biosynthesis, transport, metabolism and actions

The regulation of androgen receptor expression and only a partially formed vagina, and during
at the transcriptional and translational level is com- puberty pubic and axillary hair is scant or absent.
plex and depends on factors such as age, cell type Earlier, this syndrome of complete androgen insensi-
and tissue. Generally, androgens have a positive tivity (CAIS) was called testicular feminization (see
effect on stabilization of the receptor protein, so Chapter 3).
that androgen administration leads to inhibition of
receptor degradation and thereby to an increase in
androgen receptor protein levels. The effects of 2.6.2 Non-genomic effects of androgens
androgens on the androgen receptor mRNA are Androgens also exert rapid non-genomic effects,
the opposite. In this case, androgen administration contributing to the physiological actions (Lsel
leads to downregulation of the androgen receptor et al. 2003). Most androgenic hormone action is
mRNA by shortening of the mRNA half-life. thought to work through direct activation of DNA
Current and future research activities concentrate transcription via high affinity interactions with the
on selective androgen receptor modulators androgen receptor. Whereas genomic effects take
(SARMs), analogous to the SERMs for the estrogen hours or days to produce their actions, rapid steroid
receptor. This work leads to a general understand- effects are activated within seconds or minutes.
ing of structure-activity relationships of new phar- These non-genomic actions are not removed by
macophors of non-steroidal SARMs by structural inhibition of transcription or translation and are
modification of non-steroidal antiandrogens (see often activated by membrane-impermeant steroid
Chapter 21). conjugates. However, rapid pathways of androgen
Selective androgen receptor modulator phar- action can modulate transcriptional activity of
macophores can be classified into four categories androgen receptors or other transcription factors
so far: aryl-propionamide, bicyclic hydantoin, (Rahman and Christian 2007).
quinoline and tetrahydroquinoline analogs. In recent years, a variety of rapid non-geno-
A characteristic of these molecules is that they mic effects of sex steroids has been documented
are not substrates for aromatase or 5a-reductase. for these nuclear-oriented ligands (reviewed by
These androgen receptor ligands are intended to Simoncini and Genazzani 2003). Androgens can
act as agonists in anabolic organs (e.g. muscle and also activate transcription-independent signaling
bone) but be neutral in androgenic tissues (e.g. pathways (Heinlein and Chang 2002). Rapid effects
prostate and seminal vesicles). Other possible of androgens have been shown on calcium fluxes
molecular mechanisms related to the tissue selectivity (Guo et al. 2002) and on intracellular phosphoryl-
of SARMs include ligand-dependent changes in andro- ation cascades such as the MAP-kinase pathway
gen receptor conformation, differential interaction with (Castoria et al. 2003). Membrane effects of andro-
the promoter context of target tissue genes, and the gens have also been implicated in functional
differential recruitment of co-regulators in these tissues. responses such as rapid secretion of PSA by pro-
These new compounds are now in preclinical animal static cells (Papakonstanti et al. 2003) and the secre-
trials or already in phase I clinical trials. The hope is that tion of GnRH by pituitary cells (Shakil et al. 2002).
SARMs may be of benefit for the treatment of primary In NIH 3T3 cells, DNA synthesis is triggered after
or secondary hypogonadism, anemias, osteopenia or association between the androgen receptor and the
osteoporosis, frailty, benign hyperplasia of the prostate membrane components has occurred under the
(BPH), and may also be of help in designing a hormo- influence of nanomolar concentrations of andro-
nal male contraception method. gens. It appears that in these cells the very low
Androgen receptor defects such as deletions or density of androgen receptors is not sufficient to
inactivating mutations can profoundly alter receptor stimulate gene transcription (Castoria et al. 2003).
function. The resulting phenotype is highly variable, Androgen-stimulated gene transcription only
ranging from under virilization to testicular femin- occurs when the intracellular receptor concentra-
ization. Inactivating mutations of the androgen tion is elevated. The membranous effects of low
receptor gene in a 46,XY male with testes resulted concentrations of nuclear oriented receptors
in a female phenotype, owing to the complete lack of could represent a more general mode for steroid
all androgen activity. However, there is no uterus action in general.

27
Chapter 2: Biosynthesis, transport, metabolism and actions

Not all the membrane effects of androgens (and alternative androgen receptors can be found in
other sex steroids) are mediated by the classical eels. In eels, nanomolar concentrations of
receptor. There are several good indications that 11-ketotestosterone, for which no nuclear receptor
other steroid-binding proteins localized in the has been found, are essential for maintaining
plasma membrane are essential for signal transduc- spermatogenesis in vitro. Under these conditions
tion, but for many years the structure of these high concentrations of testosterone or DHT were
proteins could not be elucidated and therefore this inactive (Miura et al. 1991). A number of further
was not a popular subject for study. Recently an examples related to non-genomic androgen actions
alternative receptor for membrane effects of were reported more recently. Androgens acutely
progestins has been cloned (Zhu et al. 2003a). The (e.g. within approximately 30 minutes) altered the
protein has seven transmembrane domains and has frequency of the peristaltic activity of small
similarities with G-protein-coupled receptors. intestine muscle and augmented the amplitude of
Hybridization analyses have revealed that many agonist-induced contractile activity (Gonzalez-
mRNAs are present in a variety of human tissues Montelongo et al. 2011). Further studies linked
(Zhu et al. 2003b). Although a similar protein has androgen activities to calcium mobilization and
not been identified for androgens, it is known that potentially the activation of the Rho pathway.
humans can smell very small amounts of androste- Evidence has also been presented that the orphan
none (16 ene-5a-androsten-3-one) as a volatile G-coupled protein receptor GPRC6A, a widely
compound. Since only a very few isomers (but not expressed calcium- and amino-acid-sensing
testosterone) can be detected by the olfactory G-coupled receptor, transduces the non-genomic
system, it is very likely that the smell is triggered effects of testosterone and also those of other ster-
by specific membrane receptors for androstenone oids (Pi et al. 2011). Interestingly, and conversely,
in the olfactory sensory neurons (Snyder et al. the testosterone-induced rapid signaling was
1988). It is known that all olfactory receptors are reduced in prostate cancer cells with GPRC6A defi-
classical G-protein-coupled proteins, and since the ciency. Of similar interest, testosterone suppressed
alternative membrane receptor for progestins is LH secretion in wild-type mice but stimulated LH
homologous with G-coupled-receptors, it is not in GPRC6A/ null mice.
unlikely that alternative androgen receptors in the There is also considerable evidence that the non-
olfactory system have a similar structure. genomic effects of steroids are mediated via the
The dependency of spermatogenesis on high levels nuclear receptors that stimulate signaling pathways
of testosterone cannot be explained by properties of in the cytoplasm of target cells (Migliacco et al.
the classical nuclear receptor, and since the levels of 2011). Overall, though, albeit there is no longer any
testosterone required for maintaining normal sperm- doubt about the reality of non-genomic actions of
atogenesis are much higher than the saturation level androgens (Bennett et al. 2010; Grosse et al. 2011),
of the high-affinity androgen receptor, an alternative much still needs to be learned about the role
sensing system with a lower affinity has been postu- of the androgen receptor in non-gonadal tissues;
lated (Rommerts 2004). It is interesting to note that for example why certain tissues develop androgen
the alternative membrane receptor for progestins also insensitivity.
has a 10-fold lower affinity than the classical proges-
terone receptor.
2.7 Key messages
Thus, testosterone can modify the susceptibility
of T-cells to infectious diseases. Effects of testoster-  Leydig cells are the main source of testosterone
and undergo various developmental and
one on calcium mobility through cell membranes of
functional stages throughout ontogeny.
T-cells have been reported (Benten et al. 1997).  Steroidogenesis is the conversion of cholesterol
Since T-cells do not possess the classical androgen within mitochondria into pregnenolone.
receptors, this biological response also indicates the Transformation of pregnenolone into
involvement of unconventional plasma membrane testosterone occurs enzymatically in the
receptors for the expression of these androgen endoplasmic reticulum of Leydig cells.
effects. Another example of the involvement of

28
Chapter 2: Biosynthesis, transport, metabolism and actions

 Luteinizing hormone is the key regulator of  In addition to genomic effects of androgens


Leydig cell steroidogenesis. Involvement of other mediated via their nuclear receptor,
factors is likely but less well established. androgens can also exert non-genomic
 Steroid hormones diffuse through tissues without effects by interacting with cellular membrane
apparent specific transport systems. receptors.

2.8 References Bose M, Whittal RM, Miller WL, Bose


HS (2008) Steroidogenic activity of
protection assay. J Steroid Biochem
Mol Biol 55:3541
Anand-Ivell RJ, Relan V, Balvers M, StAR requires contact with
Coiffec-Dorval I, Fritsch M, Eacker SM, Agrawal N, Qian K, Dichek
mitochondrial VDAC1 and HL, Gong EY, Lee K, Braun RE
Bathgate RA, Ivell R (2006) phosphate carrier protein. J Biol
Expression of the insulin-like (2008) Hormonal regulation of
Chem 283:88378845 testicular steroid and cholesterol
peptide 3 (INSL3) hormone-
receptor (LGR8) system in the testis. Canto P, Soderlund D, Ramon G, homeostasis. Mol Endocrinol
Biol Reprod 74:945953 Nishimura E, Mendez JP (2002) 22:623635
Mutational analysis of the luteinizing Ferlin A, Arredi B, Zuccarello D,
Ariyaratne HB, Mills N, Mason JI, hormone receptor gene in two
Mendis-Handagama SM (2000) Garolla A, Selice R, Foresta C (2006)
individuals with Leydig cell tumors. Paracrine and endocrine roles of
Effects of thyroid hormone on Am J Med Genet 108:148152
Leydig cell regeneration in the adult insulin-like factor 3. J Endocrinol
rat following ethane dimethane Castoria G, Lombardi M, Barone MV, Invest 29:657664
sulphonate treatment. Biol Reprod Bilancio A, Di Domenico M, Foresta C, Bettella A, Vinanzi C,
63:11151123 Bottero D, Vitale F, Migliaccio A, Dabrilli P, Meriggiola MC,
Auricchio F (2003) Androgen- Garolla A, Ferlin A (2004) A novel
Asatiani K, von Eckardstein S, Simoni stimulated DNA synthesis and
M, Gromoll J, Nieschlag E (2003) circulating hormone of testis origin
cytoskeletal changes in fibroblasts in humans. J Clin Endocrinol Metab
CAG repeat length in the androgen by a nontranscriptional receptor
receptor gene affects the risk of 89:59525958
action. J Cell Biol 161:547556
male infertility. Int J Androl Gao HB, Tong MH, Hu YQ, Guo QS,
26:255261 Chandrashekar V, Bartke A (2007) Ge R, Hardy MP (2002)
Growth factor in Leydig cell Glucocorticoid induces apoptosis in
Bay K, Matthiesson KL, McLachlan RI, function. In: Payne A, Hardy MP
Andersson AM (2006) The effects of rat leydig cells. Endocrinology
(eds) The Leydig Cell in Health and 143:130138
gonadotropin suppression and Disease. Humana Press, Totowa, NJ,
selective replacement on insulin-like pp 263277 Ge R, Hardy MP (2007) Regulation of
factor 3 secretion in normal adult Leydig cells during pubertal
men. J Clin Endocrinol Metab Coviello AD, Matsumoto AM, development. Payne A, Hardy MP
91:11081111 Bremner WJ, Herbst KL, Amory JK, (eds) The Leydig Cell in Health and
Anawalt BD, Sutton PR, Wright Disease. Humana Press, Totowa, NJ,
Bennett NC, Gardiner RA, Hooper JD, WW, Brown TR, Yan X, Zirkin BR,
Johnson DW, Gobe GC (2010) pp 5570
Jarow JP (2005) Low-dose
Molecular cell biology of androgen human chorionic gonadotropin Ghinea N, Milgrom E (2001) A new
receptor signalling. Int J Biochem maintains intratesticular function for the LH/CG receptor:
Cell Biol 42:813827 testosterone in normal men transcytosis of hormone across
Benten WP, Lieberherr M, Sekeris CE, with testosterone-induced the endothelial barrier in target
Wunderlich F (1997) Testosterone gonadotropin suppression. organs. Semin Reprod Med 19:97101
induces Ca2+ influx via non-genomic J Clin Endocrinol Metab Gonzalez-Montelongo MC, Marin R,
surface receptors in activated T cells. 90:25952602 Gomez T, Diaz M (2011)
FEBS Lett 407:211214 Dankbar B, Brinkworth MH, Schlatt S, Androgens are powerful non-
Bertrand S, Brunet FG, Escriva H, Weinbauer GF, Nieschlag E, genomic inducers of calcium
Parmentier G, Laudet V, Robinson- Gromoll J (1995) Ubiquitous sensitization in visceral smooth
Rechavi M (2004) Evolutionary expression of the androgen receptor muscle. Steroids 75:533538
genomics of nuclear receptors: from and testis-specific expression of the Grosse A, Bartsch S, Baniahmad A
twenty-five ancestral genes to FSH receptor in the cynomolgus (2011) Androgen receptor-mediated
derived endocrine systems. Mol Biol monkey (Macaca fascicularis) gene repression. Mol Cell Endocrinol
Evol 21:19231937 revealed by a ribonuclease Jul 19 [Epub ahead of print]

29
Chapter 2: Biosynthesis, transport, metabolism and actions

Guo Z, Benten WP, Krucken J, Kishimoto M, Fujiki R, Takezawa S, subunit mutation. Fertil Steril
Wunderlich F (2002) Nongenomic Sasaki Y, Nakamura T, Yamaoka K, 90:11691174
testosterone calcium signaling. Kitagawa H, Kato S (2006) Nuclear Lsel RM, Falkenstein E, Feuring M,
Genotropic actions in androgen receptor mediated gene regulation Schultz A, Tillmann HC, Rossol-
receptor-free macrophages. through chromatin remodeling and Haseroth K, Wehling M (2003)
J Biol Chem 277:2960029607 histone modifications. Endocr J Nongenomic steroid action:
Haider SG (2004) Cell biology of 53:157172 controversies, questions, and
Leydig cells in the testis. Int Rev Klose RJ, Zhang Y (2007) Regulation of answers. Physiol Rev 83:9651016
Cytol 233:181241 histone methylation by Luetjens CM, Didolkar A, Kliesch S,
demethylimination and Paulus W, Jeibmann A, Bocker W,
Hammes A, Andreassen TK, Spoelgen
demethylation. Nat Rev Mol Cell Nieschlag E, Simoni M (2006)
R, Raila J, Hubner N, Schulz H,
Biol 8:307318 Tissue expression of the nuclear
Metzger J, Schweigert FJ, Luppa PB,
Nykjaer A, Willnow TE (2005) Role Kremer H, Mariman E, Otten BJ, Moll progesterone receptor in male
of endocytosis in cellular uptake of GW Jr, Stoelinga GB, Wit JM, non-human primates and men.
sex steroids. Cell 122:751762 Jansen M, Drop SL, Faas B, Ropers J Endocrinol 189:529539
HH, Brunner HG (1993)
Heinlein CA, Chang C (2002) The roles Maffei L, Murata Y, Rochira V, Tubert
Cosegregation of missense
of androgen receptors and G, Aranda C, Vazquez M, Clyne
mutations of the luteinizing
androgen-binding proteins in CD, Davis S, Simpson ER, Carani C
hormone receptor gene with
nongenomic androgen actions. Mol (2004) Dysmetabolic syndrome in a
familial male-limited precocious
Endocrinol 16:21812187 man with a novel mutation of the
puberty. Hum Mol Genet
aromatase gene: effects of
Huhtaniemi I (2000) The Parkes 2:17791783
lecture. Mutations of testosterone, alendronate, and
Lei ZM, Mishra S, Zou W, Xu B, Foltz estradiol treatment. J Clin
gonadotrophin and gonadotrophin
receptor genes: what do they
M, Li X, Rao CV (2001) Targeted Endocrinol Metab 89:6170
disruption of luteinizing hormone/
teach us about reproductive Manna PR, Tena-Sempere M,
human chorionic gonadotropin
physiology? J Reprod Fertil Huhtaniemi IT (1999) Molecular
receptor gene. Mol Endocrinol
119:173186 mechanisms of thyroid hormone-
15:184200
Ivell R, Anand-Ivell R (2011) Biological stimulated steroidogenesis in mouse
Lei ZM, Mishra S, Ponnuru P, Li X, Leydig tumor cells. Involvement of
role and clinical significance of
Yang ZW, Rao ChV (2004) the steroidogenic acute regulatory
insulin-like peptide 3. Curr Opin
Testicular phenotype in luteinizing (StAR) protein. J Biol Chem
Endocrinol Diabetes Obes
18:210216
hormone receptor knockout 274:59095918
animals and the effect of
Jarow JP, Chen H, Rosner TW, Metzger E, Wissmann M, Schule R
testosterone replacement therapy.
(2006) Histone demethylation and
Trentacoste S, Zirkin BR (2001) Biol Reprod 71:16051613
Assessment of the androgen androgen-dependent transcription.
environment within the human
Levalle O, Zylbersztein C, Aszpis S, Curr Opin Genet Dev 16:513517
Aquilano D, Terradas C,
testis: minimally invasive method to Migliaccio A, Castoria G, Auricchio F
Colombani M, Aranda C, Scaglia H
obtain intratesticular fluid. J Androl (2011) Analysis of androgen
(1998) Recombinant human
22:640645 receptor rapid actions in cellular
follicle-stimulating hormone
Junaidi A, Luetjens CM, Wistuba J, administration increases signaling pathways: receptor/src
Kamischke A, Yeung CH, Simoni testosterone production in association. Methods Mol Biol
M, Nieschlag E (2005) men, possibly by a Sertoli cell- 776:361370
Norethisterone enanthate has secreted nonsteroid factor. Miller WL (2007) StAR search what
neither a direct effect on the testis J Clin Endocrinol Metab we know about how the
nor on the epididymis: a study in 83:39733976 steroidogenic acute regulatory
adult male cynomolgus monkeys Lofrano-Porto A, Casulari LA, protein mediates mitochondrial
(Macaca fascicularis). Eur Nascimento PP, Giacomini L, Naves cholesterol import. Mol Endocrinol
J Endocrinol 152:655661 LA, da Motta LD, Layman LC 21:589601
King SR, Bhangoo A, Stocco DM (2008) Effects of follicle-stimulating Miura T, Yamauchi K, Takahashi H,
(2011) Functional and physiological hormone and human chorionic Nagahama Y (1991) Hormonal
consequences of StAR deficiency: gonadotropin on gonadal induction of all stages of
role in lipoid congenital adrenal steroidogenesis in two siblings with spermatogenesis in vitro in the male
hyperplasia. Endocr Dev 20:4753 a follicle-stimulating hormone beta Japanese eel (Anguilla japonica).

30
Chapter 2: Biosynthesis, transport, metabolism and actions

Proc Natl Acad Sci USA Rahman F, Christian HC (2007) Non- Snyder SH, Sklar PB, Pevsner J (1988)
88:57745778 classical actions of testosterone: an Molecular mechanisms of olfaction.
Modi DN, Shah C, Puri CP (2007) update. Trends Endocrinol Metab J Biol Chem 263:1397113974
Non-genomic membrane 18:371378 Sriraman V, Anbalagan M, Rao AJ
progesterone receptors on human Rao CV, Lei ZM (2002) Consequences (2005) Hormonal regulation of
spermatozoa. Soc Reprod Fertil of targeted inactivation of LH Leydig cell proliferation and
Suppl 63:515529 receptors. Mol Cell Endocrinol differentiation in rodent testis: a
OShaughnessy PJ, Fowler PA (2011) 187:5767 dynamic interplay between
Endocrinology of the mammalian gonadotrophins and testicular
Rommerts FFG (2004) Testosterone:
fetal testis. Reproduction 141:3746 factors. Reprod Biomed Online
an overview of biosynthesis,
11:507518
Palazzolo I, Gliozzi A, Rusmini P, Sau D, transport, metabolism and non-
Crippa V, Simonini F, Onesto E, genomic actions. In: Nieschlag E, Takamiya K, Yamamoto A, Furukawa
Bolzoni E, Poletti A (2008) The role Behre HM (eds) Testosterone: K, Zhao J, Fukumoto S, Yamashiro
of the polyglutamine tract in Action, Deficiency, Substitution, 3rd S, Okada M, Haraguchi M, Shin M,
androgen receptor. J Steroid Biochem edn. Cambridge University Press, Kishikawa M, Shiku H, Aizawa S
Mol Biol 108:245253 Cambridge, pp 138 (1998) Complex gangliosides are
essential in spermatogenesis of
Papakonstanti EA, Kampa M, Castanas E, Saudan C, Baume N, Robinson N,
mice: possible roles in the transport
Stournaras C (2003) A rapid, Avois L, Mangin P, Saugy M (2006)
of testosterone. Proc Natl Acad Sci
nongenomic, signaling pathway Testosterone and doping control.
USA 95:1214712152
regulates the actin reorganization Br J Sports Med 40 (Suppl 1):
induced by activation of membrane i21i24 Takeda K, Toda K, Saibara T,
testosterone receptors. Mol Nakagawa M, Saika K, Onishi T,
Seckl JR, Walker BR (2001)
Endocrinol 17:870881 Sugiura T, Shizuta Y (2003)
Minireview: 11beta-hydroxysteroid
Progressive development of insulin
Payne AH (2007) Steroidogenic dehydrogenase type 1 a tissue-
resistance phenotype in male mice
enzymes in Leydig cells. In: Payne specific amplifier of glucocorticoid
with complete aromatase (CYP19)
A, Hardy MP (eds) The Leydig Cell action. Endocrinology
deficiency. J Endocrinol
142:13711376
in Health and Disease. Humana 176:237246
Press, Totowa, NJ, pp 157172 Selva DM, Bassas L, Munell F, Mata A,
Tao YX, Bao S, Ackermann DM, Lei
Payne AH, Hardy MP (eds) (2007) The Tekpetey F, Lewis JG, Hammond
ZM, Rao CV (1997) Expression of
Leydig Cell in Health and Disease. GL (2005) Human sperm sex
luteinizing hormone/human
Humana Press, Totowa, NJ hormone-binding globulin isoform:
chorionic gonadotropin receptor
characterization and measurement
Penning TM, Burczynski ME, Jez JM, gene in benign prostatic hyperplasia
by time-resolved fluorescence
Hung CF, Lin HK, Ma H, Moore M, and in prostate carcinoma in
immunoassay. J Clin Endocrinol
humans. Biol Reprod 56:6772
Palackal N, Ratnam K (2000) Metab 90:62756282
Human 3alpha-hydroxysteroid Themmen AP, Martens JW, Brunner
dehydrogenase isoforms Setchell BP (2004) Hormones: what the
HG (1998) Activating and
(AKR1C1-AKR1C4) of the testis really sees. Reprod Fertil Dev
inactivating mutations in LH
aldo-keto reductase superfamily: 16:535545
receptors. Mol Cell Endocrinol
functional plasticity and tissue Setchell BP, Pakarinen P, Huhtaniemi I 145:137142
distribution reveals roles in the (2002) How much LH do the Leydig
Vandenput L, Mellstrom D, Lorentzon
inactivation and formation of male cells see? J Endocrinol 175:375382
M, Swanson C, Karlsson MK,
and female sex hormones. Biochem J Shakil T, Hoque AN, Husain M, Brandberg J, Lonn L, Orwoll E,
351:6777 Belsham DD (2002) Differential Smith U, Labrie F, Ljunggren O,
Pi M, Parrill AL, Quarles LD (2011) regulation of gonadotropin- Tivesten A, Ohlsson C (2007)
GPRC6A mediates the non- releasing hormone secretion and Androgens and glucuronidated
genomic effects of steroids. J Biol gene expression by androgen: androgen metabolites are associated
Chem 285:3995339964 membrane versus nuclear receptor with metabolic risk factors in men.
activation. Mol Endocrinol J Clin Endocrinol Metab
Prince F (2007) The human Leydig cell:
16:25922602 92:41304137
functional morphology and
developmental history. In: Payne A, Simoncini T, Genazzani AR (2003) Wikstrm AM, Bay K, Hero M,
Hardy MP (eds) The Leydig Cell in Non-genomic actions of sex steroid Andersson AM, Dunkel L (2006)
Health and Disease. Humana Press, hormones. Eur J Endocrinol Serum insulin-like factor 3 levels
Totowa, NJ, pp 7190 148:281292 during puberty in healthy boys and

31
Chapter 2: Biosynthesis, transport, metabolism and actions

boys with Klinefelter syndrome. prenatal but arrested postnatal meiotic maturation of fish
J Clin Endocrinol Metab sexual development of luteinizing oocytes. Proc Natl Acad Sci USA
91:47054708 hormone receptor knockout 100:22312236
Wu SC, Zhang Y (2009) Minireview: (LuRKO) mice. Mol Endocrinol
Zhu Y, Bond J, Thomas P (2003b)
role of protein methylation and 15:172183
Identification, classification, and
demethylation in nuclear hormone Zhu Y, Rice CD, Pang Y, Pace M, partial characterization of genes in
signaling. Mol Endocrinol Thomas P (2003a) Cloning, humans and other vertebrates
23:13231334 expression, and characterization of homologous to a fish membrane
Zhang FP, Poutanen M, Wilbertz J, a membrane progestin receptor and progestin receptor. Proc Natl Acad
Huhtaniemi I (2001) Normal evidence it is an intermediary in Sci USA 100:22372242

32
Chapter
Pathophysiology of the androgen receptor

3 Olaf Hiort, Ralf Werner, and Michael Zitzmann

3.1 Introduction 33 3.5.1 Kennedy syndrome: a pathological


3.2 Gonadal development and endocrine expansion of the AR gene CAG repeats 42
function 34 3.5.2 An animal model of the human CAG repeat
3.3 Androgens in sexual development 35 polymorphism 43
3.3.1 In sexual differentiation of the embryo 3.5.3 Ethnic differences 43
and fetus 35 3.5.4 Prostate development and
3.3.2 In male puberty and adulthood 37 malignancy 43
3.4 Androgen insensitivity in humans 38 3.5.5 Reproductive functions 44
3.4.1 Clinical findings 38 3.5.6 Bone tissue 45
3.4.2 Laboratory assessment of androgen 3.5.7 Cardiovascular risk factors 45
insensitivity 39 3.5.8 Psychological implications 46
3.4.3 Genetics 40 3.5.9 Hair growth 47
3.4.4 Functional characterization of mutants in 3.5.10 Pharmacogenetics and hypotheses 47
androgen insensitivity syndrome 41 3.6 Outlook 48
3.5 The role of CAG repeat polymorphisms of the 3.7 Key messages 49
androgen receptor in various target organs 42 3.8 References 50

3.1 Introduction thought that most 46,XY patients with presumed


The final biological steps in the cellular cascade of defects of androgen action would carry mutations
normal male sexual differentiation are initiated by in the coding regions of the AR gene. While this holds
the molecular action of androgens in androgen- true for the majority of patients with complete andro-
responsive target tissues. Lack of androgenic steroids gen insensitivity syndrome (CAIS), to an increasing
leads to defective sexual differentiation in embryos proportion patients clinically assigned as partial (PAIS)
with 46,XY karyotype; while their excess is associated or minimal androgen insensitivity syndrome (MAIS)
with virilization in 46,XX children. Furthermore, do not carry relevant mutations in the AR. Therefore
androgens lead to specific changes during male the clarification of the time- and cell-specific mechan-
puberty and are required for male fertility. isms of androgen action and the factors acting in
A key player in the translation of androgen action concert with the AR is of utmost importance. They
is the androgen receptor (AR), a nuclear transcription lead to the cell-specific modulation of androgen-
factor which can bind various androgenic steroids dependent transcription of several hundred target
as ligands and then act via differential DNA targeting genes (Holterhus et al. 2003; 2007).
and genetic control. With the elucidation of the One example is the subtle modulations of the
X-chromosomal localization and the genetic structure transcriptional activity induced by the AR that have
of the AR more than 20 years ago (Fig. 3.1), it was been assigned to a polyglutamine stretch of variable

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

33
Chapter 3: Pathophysiology of the androgen receptor

AR gene Fig. 3.1 Display of the X chromosome


with the androgen receptor (AR) gene.
p q X chromosome Exon 1 contains a variable number of CAG
repeats encoding a polyglutamine stretch
of variable length in the receptor protein.
The number of CAG repeats or length of
polyglutamine residues is inversely
associated with the transcriptional activity
Exon 1
of androgen-dependent genes, and
hence androgen effects in target tissues.
(CAG)835CAA (GGN)1027

AF1 Bipartite AF2


FQNLF NLS
2+ 2+
Zn Zn
NH2 AR protein

PolyQ PolyG
Glu 9-36 Gly 10-27

NTD DBD H LBD

length within the N-terminal domain of the receptor. ovary. After formation of the genital ridges, low
This stretch is encoded by a variable number of CAG expression of both male and female pathway genes
triplets in exon 1 of the AR gene. First observations of is initiated, independent of the genetic sex of the
pathologically elongated AR CAG repeats in patients embryo. In eutherian mammals, the presence or
with X-linked spinal and bulbar muscular atrophy absence of SRY (sex-determining region of the
(XSBMA) showing marked hypoandrogenic traits Y chromosome) is the genetic master switch that
(La Spada et al. 1991) were supplemented by partially initiates changes in the ratio of male and female
conflicting findings of clinical significance also within factors to enhance one of the two sex-determining
the normal range of CAG repeat length. The modu- pathways, leading to the differentiation of the gonad
latory effect on androgen-dependent gene transcrip- into testis or ovary (for a review see Kashimada and
tion is linear, and probably mediated by a differential Koopman 2010; Piprek 2009; Sekido and Lovell-
affinity of coactivator proteins to the encoded poly- Badge 2009). In mice Sry is expressed in XY gonads
glutamine stretch, such as ARA24 and p160 (Hsiao only in a short time-window between 10.5 and 12.5
et al. 1999; Irvine et al. 2000). As these proteins days post-coitum, with a peak at 11.5 days post-
are ubiquitously but nevertheless non-uniformly coitum in precursors of Sertoli cells, and upregulates
expressed, the modulatory effect of the CAG repeat Sry-box containing gene 9 (Sox9) expression (Hacker
polymorphism on AR target genes is most likely not et al. 1995; Bullejos and Koopman 2001; Sekido
only dependent on androgenic saturation and AR and Lovell-Badge 2008). Then Sry is downregulated
expression, but also varies from tissue to tissue. To and Sox9 is maintained by autoregulatory direct and
date, an involvement of prostate cancer risk, sperm- indirect feed-forward loops. Proper and sufficient
atogenesis, bone density, hair growth, cardiovascular Sox9 expression is necessary for recruitment of sup-
risk factors and psychological implications has been porting cell precursors (Sertoli and follicle cell pre-
demonstrated. cursors) into the Sertoli cell lineage. SOX9 can bind to
its own testis-specific enhancer (TES, testis-specific
3.2 Gonadal development and enhancer of SOX9) via the same DNA-binding motif
as SRY, and can be recruited by proteinprotein inter-
endocrine function action with SF1 (Sekido and Lovell-Badge 2008).
Normal male sexual development is dependent on SOX9 also upregulates SF1 in early testis develop-
genetic events of gonadal development as well as on ment, providing a positive feed-forward loop (Shen
endocrine pathways initiated by hormones secreted and Ingraham 2002). Another positive feed-forward
from the testes. The gonad is an organ with a bipo- loop necessary for SOX9 maintenance is upregulation
tential nature that can develop from a single primor- of fibroblast growth factor 9 (FGF9) by SOX9, which
dium, the genital ridges, either into a testis or an transduces its signals mainly by fibroblast growth

34
Chapter 3: Pathophysiology of the androgen receptor

factor receptor 2 (FGFR2) and helps to maintain (MD or paramesonephric) ducts develop in the fetus
SOX9 expression (Kim et al. 2006; 2007). Although regardless of the karyotype. Wolffian and Mullerian
early SOX9 expression is not affected in Fgf9 knock- ducts represent anlagen of the male and female repro-
out mice, maintenance of SOX9 expression is abol- ductive tracts. If testicular formation is unhindered,
ished, leading to a complete XY, female sex reversal the Leydig cells will produce testosterone (in humans
(Kim et al. 2006). at eight weeks of gestation) and the Sertoli cells
Prostaglandin D synthase (PTGDS) is another will produce anti-Mullerian hormone/Mullerian-
target of SOX9 and is required to synthesize prosta- inhibiting substance (AMH/MIS). High local concen-
glandin D2 (PGD2). Prostaglandin D2 enhances SOX9 trations of testosterone lead to the stabilization of the
nuclear transport and upregulates SOX9 expression Wolffian duct and its development into the separate
(Malki et al. 2005; Wilhelm et al. 2005); while SOX9 but connected organs, the epididymis, the vas defer-
binds to and activates the promoter of PTGDS, provid- ens and the seminal vesicles. The action of testoster-
ing another feed-forward loop of SOX9 maintenance one on Wolffian duct stabilization is mediated by the
(Wilhelm et al. 2007). In contrast to Fgf9, knock-out of androgen receptor, as shown by AR knock-out mice,
Ptgds does not disrupt gonadal development (Moniot and patients with complete androgen sensitivity
et al. 2009). Fibroblast growth factor 9 and PGD2 may syndrome (CAIS) that lack Wolffian duct derivates
also act as paracrine factors that stimulate proliferation (Yeh et al. 2002; Hannema et al. 2006; Hannema and
and differentiation of Sertoli progenitor cells. The dif- Hughes 2007).
ferentiating Sertoli cells enclose clusters of germ cells Expression of AMH induces regression of the
and form, together with the peritubular myoid cells, the Mullerian ducts in a short period between 8 and 10
testis cords, a structure that separates the germ cells weeks of gestation (Taguchi et al. 1984). In contrast,
and Sertoli cells from the interstitial cells. The Sertoli absence of AMH action leads to development of
cells are thought to also coordinate the differentiation Fallopian tubes, the uterus and the upper third of
of the interstitial steroidogenic Leydig cells and the the vagina. To exert the action of AMH, also high
peritubular myoid cells (Wilhelm et al. 2007). Steroid- concentrations of this hormone and active binding to
producing fetal Leydig cells appear 24 hours after a membrane receptor in the mesenchymal cells sur-
Sertoli cell differentiation (Habert et al. 2001; Yao rounding the Mullerian ducts are necessary. There-
et al. 2002). Desert hedgehog (DHH), a Sertoli-derived fore, reduced excretion of AMH due to lowered
signaling molecule, has been shown to play an import- numbers of Sertoli cells is responsible for partial
ant role in fetal Leydig cell differentiation (Yao et al. uterus formation seen clinically in sex determination
2002). Ectopic activation of the Hedgehog signaling disorders (Ostrer 2008).
pathway in fetal ovaries leads to a transformation of
SF1-positive somatic cells into functional fetal Leydig
cells producing androgens and INSL3 (Barsoum et al. 3.3 Androgens in sexual development
2009; Barsoum and Yao 2010). In mammals, two dis- 3.3.1 In sexual differentiation of the
tinct populations of Leydig cells appear sequentially
during testis development: fetal Leydig cells and adult embryo and fetus
Leydig cells. Fetal Leydig cells start to produce andro- The prostate develops from the urogenital sinus
gens around 13 days post coitum in mice, leading to (UGS), a ventral urinary compartment formed after
the masculinization of the male urogenital system separation of the digestive and urinary tract by the
(OShaughnessy et al. 2006). In mice, in contrast to urorectal septum. The sexually dimorphic develop-
adult Leydig cells, fetal Leydig cells do not express 17b- ment of the UGS is dependent on androgens secreted
HSD, and secrete androstenedione, which is converted by the Leydig cells of the developing testis (for a
within the seminiferous tubules, most likely by Sertoli review see Marker et al. 2003; Meeks and Schaeffer
cells, into testosterone (OShaughnessy et al. 2000). The 2011). Androgen-dependent development of the pro-
development of fetal Leydig cells is independent of state was demonstrated by de novo induction of
androgens up to postnatal day five, as evidenced by prostatic buds from male and female urogenital
AR knock-out mice (OShaughnessy et al. 2002). sinuses of rats using organ culture methods (Lasnitzki
In early gestation, two pairs of unipotential ducts, and Mizuno 1977; 1979; Takeda et al. 1986). Although
the Wolffian (WD or mesonephric) and Mullerian prostate development begins with the development of

35
Chapter 3: Pathophysiology of the androgen receptor

epithelial buds, the AR is expressed at these stages upregulated by testosterone. Dependency on andro-
only in the surrounding mesenchyme (Takeda et al. gens was confirmed by a five-fold decreased expres-
1986), indicating that paracrine androgen-dependent sion of SHH in the absence of DHT in in-vitro organ
signals may mediate bud formation. Androgen culture of UGS (Podlasek et al. 1999). Although the
dependency of prostate development is also shown DHT-dependent development of the prostate is estab-
by the lack of a prostate in AR knock-out mice and lished downstream, effectors of androgen signaling
in humans with CAIS (Zhou 2010). In addition, some mediating the fetal differentiation of the prostate have
46,XX DSD (disorders of sexual development) been poorly defined.
patients suffering from CAH with intrauterine expos- Up to the seventh week of gestation in humans and
ure to high androgen levels have developed prostatic day of embryogenesis (E) 16.5 in mice, the external
tissue (Klessen et al. 2005; da Costa Rose Paulino genitalia of male and female embryos are morpho-
et al. 2009). Since the concentration of testosterone logically identical and consist of bipotential anlagen,
in the fetal circulation is relatively low, target cells in the genital tubercle (GT), the urogenital folds and the
the prostate and the external genitalia express 5a- bilateral genital swellings. Under influence of DHT,
reductase type 2, an enzyme that converts testoste- the GT develops into a penis and the genital swellings
rone into the 10-fold more potent androgen DHT fuse and grow into a scrotum. With the growing penis,
(Deslypere et al. 1992). Urogenital sinus organ culture a ventral urethral groove appears, with lateral urethral
experiments with testosterone or DHT led to the folds which fuse, and, together with a distal canalizing
conclusion that the effect of testosterone is mediated urethral plate, forms the urethra. In females the GT
by conversion to DHT (Lasnitzki and Mizuno 1977). develops into a clitoris, and the genital swellings and
On the other hand, inhibition of 5a-reductase by in- urogenital folds remain separate and form the labia
utero application of the 5a-reductase inhibitor finas- majora and labia minora (for a review see Yamada
teride led to a significant decrease in prostate size in et al. 2003; 2006). Like the prostate, the GT also
rats (Imperato-McGinley et al. 1992). In humans with develops through epithelialmesenchymal interactions
5a-reductase type 2 deficiency and in mice with (Murakami and Mizuno 1986; Kurzrock et al. 1999),
targeted disruption of 5a-red 2, the UGS is specified but the factors mediating the masculinization are not
as a prostate but prostate growth and development are yet known. Recently, the critical time-window for
greatly reduced (Andersson et al. 1991; Mahendroo androgen action on genital masculinization has been
et al. 2001). defined to embryonic stage E15.5 in mice. At this time
Several AR-dependent target genes have been point the AR is expressed in the mesenchyme as well
described in fetal prostate development. Nkx3.1 is a as in the urethral plate epithelium (UPE) (Miyagawa
homeobox transcription factor expressed early in the et al. 2009). Targeted disruption of mesenchymal
developing prostate buds and its expression is andro- versus epithelial AR showed a demasculinization in
gen dependent. Castration of mice leads to immediate male mesenchymal AR knock-out mice, indicating
ablation of Nkx3.1 expression (Bieberich et al. 1996). that proper mesenchymal AR signaling is necessary
Nkx3.1 is a direct target of the AR, and its androgen- for GT development. In epithelial AR knock-out mice
dependent transcription is mediated by androgen no effect on GT development could be detected (Miya-
response elements (AREs) in the 30 UTR of the gene gawa et al. 2009). Gene expression analysis of GT from
(Thomas et al. 2010). Targeted disruption of Nkx3.1 E15.5 mice revealed increased expression of the Wnt
in mice leads to defects in ductal morphology and inhibitory genes Dickkopf (DKK) and secreted frizzled
secretory protein production (Bhatia-Gaur et al. related protein 1 (SFRP1) in females, demonstrating
1999). The homeobox genes Hoxa13, Hoxb13 and a sexually dimorphic expression of Wnt signaling
Hoxd13 are expressed during UGS development and genes in the GT (Nishida et al. 2008; Miyagawa et al.
in the developing prostate (Zeltser et al. 1996; Warot 2009). A sexually dimorphic Wnt/b-catenin signaling
et al. 1997). A slight testosterone-dependent upregu- enhanced in the male GT was observed in a BatGAL
lation of these Hox genes was observed in organ Wnt/b-catenin indicator mouse line. Forty percent of
cultures of the ventral prostate of newborn rats, but conditional b-catenin loss of function mutants failed
not in the lateral prostate (Huang et al. 2007). Sonic to develop a proper prepuce and midline formation;
hedgehog (SHH; see below) is expressed in the uro- while female b-catenin gain of function mutants
genital epithelium (UGE) of prostatic buds and is showed enlarged external genitalia with a well-developed

36
Chapter 3: Pathophysiology of the androgen receptor

prepuce similar to androgen-treated females, but no The morphogenetic result of these specific actions
enlarged anogenital distance (Miyagawa et al. 2009). of androgens is the irreversible virilization of the
These results made b-catenin a good candidate as a external male genitalia. In humans, this process is
potentially downstream effector of androgen signal- terminated in the 12th week of gestation. Hence,
ing involved in external genitalia masculinization that incomplete masculinization, for example incomplete
may act by the canonical b-catenin/Tcf/Lef pathway closure of the midline (hypospadias) during the
or as an AR coactivator (Song et al. 2003). sensitive window between the 7th and the 12th week
Sonic hedgehog is a secreted growth factor cannot be overcome even by high doses of androgens
expressed in the urethral plate epithelium; whereas at later stages of development. This fact may seem
its receptor patched1 (PTC1), or its target the tran- trivial, but it clearly indicates that the genomic pro-
scription factor glioblastoma 1 (GLI1), is expressed in grams provided by the androgen target tissues must
adjacent mesenchyme (Miyagawa et al. 2011). The have undergone comprehensive and definitive alter-
role of Shh in epithelialmesenchymal interactions ations in parallel to the ontogenetic process of external
and androgen-independent outgrowth of the GT has virilization.
been acknowledged, and Shh knock-out mice display
complete GT agenesis (Haraguchi et al. 2001; Perriton
et al. 2002; Seifert et al. 2010). Recently, Miyagawa 3.3.2 In male puberty and adulthood
et al. (2011) showed by conditional Gli and Shh Puberty is a period of development with two main
knock-out mice that Hedgehog signaling also affects processes: namely adrenarche and gonadarche. Adre-
sexually dimorphic mesenchymal differentiation of narche describes the increasing androgenic steroid
the GT. Conditional Gli2/ mice showed a com- secretion from the adrenals, which is usually observed
plete loss of Hedgehog responsiveness within the between 8 and 10 years in both girls and boys, and
mesenchyme adjacent to the urethral plate epithelium is limited to primates (Plant and Witchel 2006).
in a lacZ reporter line associated with GT hypoplasia Gonadarche describes the enlargement of the gonads
and defects in the ventral midline that are even more with formation of sex steroids and the induction of
prominent in the male than in the female. Demascu- fertility. Thus, only gonadarche is fundamental to the
linization of the male GT was also associated with process of puberty and reproduction. Gonadarche is
decreased sexually dimorphic gene expression in the triggered by increasing pulsatility of GnRH secretion
GT. Interestingly, female Gli2/ mice do not mas- leading to a release of the pituitary hormones LH
culinize with androgen treatment, indicating a require- and FSH. Luteinizing hormone in turn is responsible
ment of Gli2 for GT masculinization. On the other for increasing testosterone synthesis in the testes via
hand, other androgen effects like enlargement of direct stimulation of the Leydig cells (Plant 2006).
the anogenital distance are not affected. Conditional In males, puberty is clinically evident with the
gain of function mutants for Shh signaling lead to a enlargement of testes and penis. Testicular volume
partial masculinization of the female GT by prepuce increases from 1 to 2 cm3 prepubertally to reach 20
hypertrophy, but no fusion of the urethral folds 30 cm3 in adulthood. In addition to changes in second-
was observed. In contrast to b-catenin signaling, Shh ary hair, and genital changes, increasing testosterone
signaling appears not to be a downstream target, concentrations produce other changes in most tissues
but may act cooperatively with androgen signaling of the body (Welsh et al. 2009). The larynx increases in
(Miyagawa et al. 2011). To date, androgen-dependent size and the voice deepens. Also bone mass and muscle
effectors of masculinization of the external genitalia strength increase, a growth spurt occurs, the erythro-
other than the androgen receptor have been poorly cyte cell mass increases, the skin thickens, and hair
defined. Participation of Wnt and Hedgehog signaling growth on the trunk is enhanced; androgenic hair
pathways in sexually dimorphic development of the recession may be induced as well. Furthermore, the
GT has been demonstrated. Neither targeted deletion internal sex organs such as prostate and seminal ves-
nor gain of function mutations in these pathways could icles enlarge. Sex steroids, and specifically testosterone,
mimic complete sex reversal as could the absence of may alter behavior, and central nervous effects include
androgens or the AR. Thus other downstream effectors stimulation of sexual libido and aggressiveness.
of androgen action must exist, whose identity and role In conjunction with FSH, testosterone is an essen-
in genital development still remain elusive. tial endocrine factor for spermatogenesis in male

37
Chapter 3: Pathophysiology of the androgen receptor

Fig. 3.2 Clinical grades of ambiguous


genitalia. Virilization is diminished from
grade 1 towards grade 5. According to
Sinnecker et al. 1997.

1 2 3 4 5

mammals which acts directly on the germinal epithe- However, at the time of puberty, the specific clin-
lium via the AR. During hormonal quiescence in ical appearance of AIS is much more striking. The
prepuberty, germ-cell proliferation is arrested until timing of puberty apparently follows a female pattern
the juvenile phase. Testosterone alone can induce (Papadimitriou et al. 2006). A main feature of puber-
spermatogenesis if administered during this period tal development is the feminization that occurs in
(Marshall et al. 1984). However, quantitative main- almost all patients with CAIS and PAIS with gonads
tenance of the spermatogenic process cannot be in situ. Patients with CAIS show a female puberty
achieved by testosterone alone, but needs the support- with normal onset of breast development. This is
ive action of FSH (see Chapter 6). different in all other forms of 46,XY disorders of sex
Current concepts state that mammary gland development, where only virilization might be seen at
development is mainly dependent on estrogens; the time of puberty (Werner et al. 2010).
numerous studies primarily using mouse models have Most strikingly, patients with 5a-reductase defi-
revealed that estrogen is a critical requirement for ciency or with 17b-hydroxysteroid dehydrogenase
ductal elongation during puberty, whereas progester- deficiency mostly demonstrate a strong surge in tes-
one and prolactin signaling are crucial during preg- tosterone and DHT, leading to marked virilization
nancy and lactation (LaMarca and Rosen 2008). The and sometimes to gender change if the individual
role of the AR in breast development is unclear. had been raised as a female (Hiort et al. 2003).
In AIS, the testes enlarge and histologically show
3.4 Androgen insensitivity in humans Leydig cell hyperplasia as well as a Sertoli-cell-only
pattern without evidence for spermatogenesis. An
3.4.1 Clinical findings important feature of CAIS is the insufficiency of
Defective androgen action caused by cellular resistance sexual hair growth. In some patients with CAIS, pubic
to androgens causes the androgen insensitivity syn- hair is reported, but this may appear thinner than
drome (AIS) (Quigley et al. 1995; Hiort et al. 1996). normal and may not reach Tanner stage 5 (Sinnecker
The end-organ resistance to androgens results in a wide et al. 1997).
clinical spectrum of defective virilization of the external In PAIS, a mixture of virilization and feminization
genitalia in 46,XY individuals (Fig. 3.2). In AIS, testicu- may occur. This leads to phallic and testicular
lar function is apparently normal. However, affected enlargement with sexual hair growth on one side,
individuals will have a clinical phenotype that is con- but breast development on the other side. Puberty
sistent with a varying degree of presumable androgen may not fully develop, although laboratory values
deficiency, but testis may be of low-normal size and for testosterone are measured in the adult reference
histology in childhood, and Muller structures (oviducts, range (Ahmed et al. 2011). The voice may not deepen
uterus and upper third of the vagina) are absent due to appropriately and acne is scarce. Body proportions
normal AMH synthesis and action. In this regard, at tend to be female or intermediate. Gynecomastia
clinical investigation AIS in childhood is a phenocopy is present in almost all patients with PAIS, as we
from disorders of androgen synthesis not affecting reported in a recent survey of 15 individuals assigned
adrenal function, such as 5a-reductase deficiency or to male sex (Steltenkamp and Hiort 2007). These
17b-hydroxysteroid dehydrogenase deficiency. features make clinical assessment of androgen

38
Chapter 3: Pathophysiology of the androgen receptor

insensitivity much easier during puberty and there- impaired pituitary feedback mechanism in androgen
after than during infancy and childhood. insensitivity syndrome, the postnatal surge of gona-
However, within families, the phenotype may vary dotropins and testosterone is missing in CAIS, but
considerably between MAIS and PAIS, even with the may be normal in PAIS. Anti-Mullerian hormone and
same underlying molecular abnormality (Rodien et al. inhibin B measurements were reported as normal or
1996; Holterhus et al. 2000). even elevated, because the inhibited AR expression
The fate of the gonads in CAIS is a point of accounts for the absence of AMH repression during
current debate. While the gonads in patients with testis development (Boukari et al. 2009). Rey et al.
PAIS and female sex assignment are removed to avoid (1999) reported AMH as a valuable marker of testicu-
possible virilization at the time of puberty, this point lar differentiation. While AMH was low in testicular
is not valid in CAIS patients. In the past, their gonads dysgenesis, values were within the normal range in
were often removed at the time of diagnosis, at latest disorders of androgen biosynthesis or action. Inhibin
at puberty, due to a possible increased risk for malig- B as a Sertoli cell marker showed a similar response. It
nancy. This has to be weighed against the invasive was measured low in anorchia or gonadal dysgenesis
and psychologically stressing nature of this proced- and normal or elevated in disorders of androgen
ure, which inevitably leads to the need for hormone biosynthesis or AIS compared to normal 46,XY boys
replacement therapy to induce puberty and maintain (Kubini et al. 2000).
adult health in these patients. Mostly, tumors of the At the time of puberty, gonadotropins rise nor-
gonads occur as type II germ cell tumors, namely mally. In due course of puberty, FSH might become
classic seminoma in the testis or dysgerminomas in elevated above the normal range because of secondary
the dysgenetic gonads of disorders of sex determin- testicular atrophy. Luteinizing hormone is in the
ation. Apparently, those disorders that are associated upper normal male reference range or even elevated.
with normal testicular development and histology Subsequently, a surge of testosterone is seen that does
often depict a maturation delay rather than a pre- not correspond to external masculinization, thus
neoplastic change. Therefore, the tumor risk in CAIS giving rise to the diagnosis of androgen insensitivity.
is seemingly lower than in other forms of DSD (Cools Usually, testosterone levels are in the upper male
et al. 2011). reference range. An androgen sensitivity index has
This leads to a novel view of leaving the gonads in been postulated as the mathematical product of LH
situ with the possibility that individuals with CAIS and testosterone serum values (Aiman et al. 1979) and
synthesize their own sex steroids with the very special is helpful in the discrimination of adult patients with
CAIS hormonal profile. If the gonads are maintained MAIS as a cause of male infertility, although it does
after puberty, however, the patients should be aware not differentiate AIS from other causes of hormonal
of the lack of long-term outcome data and they imbalance such as estrogen insufficiency in MAIS
should be involved in a regular screening program (Hiort et al. 2000). Thus, the higher the androgen
with imaging of the gonads as well as regular labora- sensitivity index, the more likely is an abnormality
tory assessment. of the AR gene.
The patients have strong features of feminization,
3.4.2 Laboratory assessment of androgen although estradiol levels are not elevated above the male
reference range. It can be postulated that either a high
insensitivity degree of intracellular aromatization of testosterone to
The determination of laboratory values relies on age- estradiol or the loss of repression of feminizing genes is
dependent reference values during infancy and child- responsible for this finding (Doehnert et al. 2011).
hood and is difficult to assess in androgen insensitivity. Before the time of molecular characterization of
Bouvattier et al. (2002) reported that in children with the AR gene, analysis of specific androgen binding in
CAIS, LH levels were comparably low during mini- genital skin fibroblasts of patients had been a major
puberty (measurement at day 30); while, in PAIS, they diagnostic procedure. In the age of molecular genet-
rose to a normal level, which corresponds to a refer- ics, this type of analysis has been abandoned because
ence range of Tanner 45. Also, the testosterone levels it requires the invasive method of a genital skin
were low in CAIS infants, but stimulated properly biopsy to gain the cells to be investigated. As fewer
in PAIS. The authors concluded that, due to the and fewer patients are operated on, biopsies are often

39
Chapter 3: Pathophysiology of the androgen receptor

not available. Furthermore, androgen binding studies The AR shares its particular modular composition
were able to detect only mutations of the AR located of three major functional domains with the other
in the ligand-binding part of the receptor, while steroid hormone receptors. A large N-terminal
mutations affecting the DNA-binding region of the domain precedes the DNA-binding domain (DBD)
AR usually have normal androgen binding capacities, that is connected by a short hinge region to the
but lack transactivational function. C-terminal LBD (Fig. 3.1). The N-terminal domain
An androgen sensitivity test was described on the (NTD) is encoded by the first exon and contains the
basis of the response of serum levels of SHBG after strong ligand-independent transactivation domain
intake of stanozolol, a synthetic anabolic steroid. In named activation function 1 (AF1) (Simental et al.
normal probands, serum levels of SHBG decline to 1991; Jenster et al. 1995; Chamberlain et al. 1996).
about 50% of the initial value five to eight days after The N-terminal domain is proposed to be a highly
the start of stanozolol treatment (Sinnecker et al. flexible and disordered domain that adopts a higher
1997). In AIS, this response is diminished, correlat- structure by induced folding when in contact with
ing somewhat with the phenotypic degree of under- transcriptional regulators such as the basal transcrip-
masculinization. While the SHBG test can be tion factor TFIIF (RAP74) (Lavery and McEwan 2006;
valuable as the only available in-vivo test of androgen 2008a; 2008b). The N-terminal domain harbors two
insensitivity, it has several pitfalls. Namely, stanozo- polymorphic repeats, a polyglutamine repeat of 936
lol has been taken off the regular market due to its residues and a polyglycine repeat of 1027 glycine
high potential for misuse as a doping agent and its residues, and its repeat length affects AR transactiva-
liver toxicity. Second, due to the endogenous testos- tion (Chamberlain et al. 1994; Kazemi-Esfarjani et al.
terone surge during the first months of life, this test is 1995; Wang et al. 2004; Werner et al. 2006a). In
not sensitive during this important period for diag- contrast to the N-terminal domain, the DNA-binding
nosis and management of the child with presumed domain and LBD are highly structured domains, and
AIS. And third, the test might fail in patients with their crystal structure has been resolved (Matias et al.
somatic mosaicism of a mutation in the AR gene 2000; Sack et al. 2001; He et al. 2004; Shaffer et al.
(Sinnecker et al. 1997; Holterhus et al. 1999).There- 2004). The DNA-binding domain is encoded by exon
fore at present it should be proposed only for patients 2 and 3 and consists of two zinc fingers followed by a
with unclear disorders of androgen action over the C-terminal extension. The first zinc finger contains
age of one year. the so-called P-box, an a-helix that makes the base-
specific contact with the major groove of the DNA,
while the second zinc finger containing the so-called
3.4.3 Genetics D-box is involved in DNA-dependent dimerization
The AR belongs to the intracellular family of struc- (Claessens et al. 2008). All classical steroid receptors
turally related steroid hormone receptors. Transcrip- (AR, progesterone receptor (PR), mineralocorticoid
tional regulation through the AR is a complex multi- receptor and glucocorticoid receptor) can bind to
step process involving androgen binding, conforma- the same hormone-responsive element (Mangelsdorf
tional changes of the AR protein, receptor phosphor- et al. 1995), but, in addition, AR can bind to selective
ylation, nuclear trafficking, DNA binding, cofactor AREs not bound by the glucocorticoid receptor
interaction and finally transcription activation. It is (Claessens et al. 2001; 2008). The hinge region,
now more than 20 years ago that the human AR gene encoded in part by exon 3 and 4, is defined as a
was cloned by several groups and mapped to Xq1112 flexible linker between the DNA-binding domain
(Chang et al. 1988; Trapman et al. 1988; Lubahn et al. and the LBD, and contains the bipartite nuclear local-
1988; 1989; Tilley et al. 1989). It spans approximately ization signal (NLS) (Jenster et al. 1992; 1993; Zhou
90 kb and comprises 8 exons, named 18 or AH. et al. 1994; Cutress et al. 2008). The C-terminal exten-
Transcription of the AR gene and subsequent splicing sion of the second zinc finger is also part of the hinge
usually results in distinct AR-mRNA populations in and involved in specificity of selective AREs (Haelens
genital fibroblasts. Translation of the mRNA into the et al. 2003; Schauwaers et al. 2007). The LBD is
AR protein usually leads to a product migrating at encoded by exon 48 and contains 12 a helices and
about 110 kDa in Western immunoblots comprising 4 small b-strands that assemble into a three-layer,
between 910 and 919 amino acids. sandwich-like structure with a central ligand binding

40
Chapter 3: Pathophysiology of the androgen receptor

pocket (He et al. 2004). Agonist binding leads to a function (Hiort et al. 1996). Mutations within the
repositioning of helix 12 and seals the ligand binding LBD may alter androgen binding but may in addition
pocket like a mouse trap (Moras and Gronemeyer influence dimerization due to disruption of NC-
1998). Thereby a hydrophobic cleft is formed called terminal structural interactions. Mutations within
activation function 2 (AF2) that interacts with LxxLL- the DNA-binding domain can affect receptor binding
and FxxLF-like motifs of co-regulators as well as with to target DNA. To date, only one patient with com-
the 23FQNLF27 motif in the N-terminal domain of plete AIS without an AR gene mutation but with
the AR. Additional functional subdomains could be clear experimental evidence for an AR-coactivator
identified by in-vitro investigation of artificially trun- deficiency as the only underlying molecular mechan-
cated, deleted or point-mutated ARs (see http:// ism of defective androgen action has been reported
androgendb.mcgill.ca/). Upon entering target cells, (Adachi et al. 2000). Cofactors of the AR will pre-
androgens interact very specifically with the ligand- sumably play a pivotal role in the understanding
binding pocket of the AR. This initiates an activation of phenotypic variability in AIS. So far, only a few
cascade with conformational changes and nuclear mechanisms contributing to the phenotypic diversity
translocation of the AR. Prior to receptor binding to in AIS have been identified in affected individuals.
target DNA, homodimerization of two AR proteins A striking phenotypic variability in a family with
occurs in a ligand-dependent manner. This is medi- partial AIS has been attributed to differential expres-
ated by distinct sequences within the second zinc sion of the 5a-reductase type 2 enzyme in genital
finger of the DNA-binding domain as well as through fibroblasts (Boehmer et al. 2001). Another mechan-
specific structural NC-terminal interactions. The ism may be the combination of varying androgen
AR-homodimer binds to hormone responsive elem- levels during early embryogenesis and partially inacti-
ents which usually consist of two palindromic (half- vating mutations of the LBD (Holterhus et al. 2000).
site) sequences within the promoter of androgen- Moreover, post-zygotic mutations of the AR gene
regulated genes. Through chromatin remodeling, resulting in a somatic mosaicism of mutant and wild-
direct interaction with other transcription factors type AR genes can contribute to modulation of
and specific coactivators and co-repressors, a steroid- the phenotype. This can result in a higher degree of
receptor-specific modulation of the assembly of the virilization than expected from the AR mutation
preinitiation complex is achieved, resulting in specific alone because of the expression of the wild-type AR
activation or repression of target gene transcription. in a subset of somatic cells. Due to the fact that at least
More than 800 different mutations have been one-third of all de-novo mutations of the AR gene
identified in AIS to date (http://androgendb.mcgill. occur at the post-zygotic stage, this mechanism is not
ca/). Extensive structural alterations of the AR can only important for phenotypic variability in AIS
result from complete or partial deletions of the AR but also crucial for genetic counseling. All kinds of
gene. Smaller deletions may introduce a frame shift mutations, from missense to nonsense as well as
into the open reading frame, leading to a premature frame shift and splice-site mutations have been iden-
stop codon downstream of the mutation. Similar tified in AIS patients with somatic mosaicism, and
molecular consequences arise from the direct intro- are associated with both PAIS and CAIS (Hiort et al.
duction of a premature stop codon due to point 1998; Holterhus et al. 1999; 2001; Kohler et al. 2005).
mutations. Such alterations usually lead to severe Nevertheless, a substantial fraction of mosaic muta-
functional defects of the AR and are associated with tions may escape detection because of an unequal
CAIS. Extensive disruption of the AR protein struc- distribution of mutant vs. wild-type AR in various
ture can also be due to mutations leading to aberrant tissues of an individual. This may result in a low
splicing of the AR mRNA (Hellwinkel et al. 1999; percentage of leukocytes with mutant AR that is
2001). However, as aberrant splicing can be partial undetectable by conventional Sanger sequencing.
and thus enable expression of the wild-type AR, the
AIS phenotype is not necessarily CAIS but may also 3.4.4 Functional characterization of
present with PAIS (Hellwinkel et al. 2001). The most
common molecular defects of the AR gene are mis- mutants in androgen insensitivity syndrome
sense mutations. They may either result in CAIS or in Diagnosis of AIS is confirmed in most cases by
PAIS because of complete or partial loss of AR molecular genetic analysis and the detection of

41
Chapter 3: Pathophysiology of the androgen receptor

aberrations in the AR gene. The impact of many Employing genome-wide analysis in genital skin
mutations on AR function has already been proven fibroblasts from patients with complete AIS com-
(http://androgendb.mcgill.ca/), but the effect of new pared to 46,XY male reference cells, distinct andro-
mutations on protein activity has to be established. gen-regulated gene profiles could be assessed which
Androgen receptor mutations are often recreated in correlated to the genital phenotype (Holterhus et al.
mammalian expression vectors and cotransfected into 2003). In peripheral blood monocytes a different, but
an AR-negative mammalian cell line together with an also distinct androgen-dependent expression, was
androgen-responsive reporter gene. These assays are verified (Holterhus et al. 2009). These patterns were
sensitive for many functions of the AR including independent of the current endocrine status, but
hormone binding, protein stability, nuclear transport, rather reflected an embryonal androgen influence on
DNA binding, or transcriptional activation. Impaired cell development. This proves that androgens lead to a
AR function results in reduced reporter gene expres- fingerprint of gene expression patterns, which are
sion when compared to a wild-type AR construct. distinct for cell-type, possibly mirroring genital
Mutations associated with complete or severe forms phenotype in all individuals. Detection of apolipopro-
of AIS often demonstrate a good genotypephenotype tein D as a highly specific androgen-regulated gene in
correlation in these functional studies. This holds genital skin fibroblasts in response to incubation with
especially true for mutations that disrupt the highly DHT (Appari et al. 2009) is promising. Its use as a
structured LBD or DNA-binding domain, leading possible biomarker of androgen insensitivity, espe-
to severely impaired ligand or DNA-binding. How- cially in those patients where a mutation in the AR
ever, for mutations associated with MAIS or less gene cannot be detected, remains to be proved.
severe forms of PAIS, experimental evidence is often
lacking or scarce.
This is due to the fact that, through different 3.5 The role of CAG repeat
mutations within the AR, the reduced activation of polymorphisms of the androgen
androgen-dependent reporter genes can be highly
variable. It depends on the type of mutation and its receptor in various target organs
localization within the gene, but also on the promoter 3.5.1 Kennedy syndrome: a pathological
of the experimental target gene (Werner et al. 2006b).
Furthermore, the conformational changes that will expansion of the AR gene CAG repeats
be induced by ligand-binding or chaperone inter- Kennedy syndrome or X-linked spinal and bulbar
action, the post-translational modifications, as well muscular atrophy (XSBMA) is a rare inherited neu-
as the trafficking within the cell, and further molecu- rodegenerative disease characterized by progressive
lar interaction may influence target gene activation. neuromuscular weakness caused by a loss of motor
In particular the cell-dependent expression of coacti- neurons in the brain stem and spinal cord. Disease
vators and co-repressors of the AR might modulate onset, developing in the third to fifth decade of life, is
the cellular androgen response in a very distinct and likely to be preceded by muscular cramps on exertion,
highly specific manner (Bebermeier et al. 2006). tremor of the hands and elevated muscle creatine
Therefore, the use of different reporter genes and cell kinase (Kennedy et al. 1968). The initial description
lines may be necessary to prove the pathogenicity of one of the individuals affected with Kennedy syn-
of a new variation. Detailed functional analysis of drome also contains information about gynecomastia,
distinct features of the mutant AR, such as ligand- a hypoandrogenic symptom (Kennedy et al. 1968).
binding assays, NC-terminal interaction assays, Subsequent reports emphasized the presence of symp-
EMSA (electrophoretic mobility shift assay) or toms indicating the development of androgen insensi-
nuclear localization assays may provide a more pro- tivity in men with XSBMA, exhibiting varying degrees
nounced effect on AR function, and support the of gynecomastia, testicular atrophy, disorders of
pathogenic role of a mutation. spermatogenesis, elevated serum gonadotropins and
The elucidation of androgen-regulated genes is also diabetes mellitus (e.g. Arbizu et al. 1983). Thus,
of utmost importance to understand the androgen- the AR was regarded as a candidate gene for XSBMA,
dependent cell regulation and, hence, the physiology and the expansion of the polyglutamine repeat within
and pathophysiology of sexual differentiation. the N-terminal region was further recognized as the

42
Chapter 3: Pathophysiology of the androgen receptor

cause (La Spada et al. 1991). The longer the CAG subpopulations (Kittles et al. 2001). In healthy Cau-
repeat in the AR gene, the earlier the onset of the casians the mean number of CAG repeats is 21 to 22
disease is observed and the more severe are the symp- (Edwards et al. 1992; Platz et al. 2000), while in East
toms of hypoandrogenicity (Doyu et al. 1992; Choong Asians a mean of 2223 triplets is found (Hsing et al.
and Wilson 1998; Mariotti et al. 2000; Dejager et al. 2000a; Platz et al. 2000; van Houten and Gooren 2000;
2002). The absence of any neuromuscular deficit or Wang et al. 2001). These differences can possibly be
degeneration in patients with complete androgen held responsible for some variations in androgen-
insensitivity (CAIS) (Quigley et al. 1995) suggests that dependent diseases and features which are observed
neurological deficits in XBSMA are not caused by a among different ethnicities; for example beard growth
lack of androgen influence but rather by a neurotoxic or rate of prostate cancer.
effect associated with the pathologically elongated
number of CAG repeats, which causes irregular pro- 3.5.4 Prostate development
cessing of the AR protein and accumulation of end-
products (Abdullah et al. 1998). and malignancy
The prostate is an androgen-regulated organ (see
Chapter 13). A substantial difference exists in the
3.5.2 An animal model of the human incidence of prostate cancer between ethnic groups,
CAG repeat polymorphism with African Americans having a 20- to 30-fold
A humanized mouse model of the AR gene (CAG)n higher incidence than East Asians (Hsing et al.
directly assessed the functional significance of the AR 2000b). Such disparity cannot be explained entirely
gene (CAG)n: the mouse AR was converted to the by screening bias in different populations. Also after
human sequence by germline gene targeting, introdu- multiple adjustments for ethnic and screening differ-
cing alleles with 12, 21 or 48 (CAG)n. The three ences, a significant contrast in incidence rates
humanized AR mouse lines revealed markedly dif- between African Americans, Caucasians and Asians
ferent seminal vesicle weights indicating androgen is found (Ross et al. 1998; Platz et al. 2000).
effects, despite similar serum testosterone concentra- It can be assumed that a polymorphism of the AR
tions. Molecular analysis of AR-dependent target gene with the capacity to modulate androgen effects has an
expression demonstrated Nkx3.1 and clusterin influence on the fate of malignant cells in the prostate.
mRNAs to be modulated by the number of (CAG)n Thus, with shorter CAG repeats, an earlier onset of
in the prostate probasin. Crossed with transgenic- the disease would be observed, as well as an associ-
adenocarcinoma-of-mouse-prostate mice, genotype- ation with aggressiveness of the tumor. Investigation
dependent differences in prostate cancer prevalence of a younger study group would then lead to the
were observed (Albertelli et al. 2006). This confirms supposition of an increased risk of developing pro-
findings of AR gene (CAG)n expression in human state cancer. While this would hold true for a specific
prostate cancer cells (Coutinho-Camillo et al. 2006). younger age group, it is likely that the effect cannot be
observed when older men are also involved since
the overall incidence of prostate cancer is high. Strati-
3.5.3 Ethnic differences fication for lifestyle factors and multidimensional
The normal range of CAG repeats is 9 to 37, and matching of controls in a sufficient number of sub-
follows a normal, slightly skewed distribution towards jects is a prerequisite for relevant investigations: this
the higher number of triplets (Edwards et al. 1992; is best met by eight studies (Giovannucci et al. 1997;
Platz et al. 2000; Hsing et al. 2000a; Kuhlenbumer Stanford et al. 1997; Platz et al. 1998; Correa-Cerro
et al. 2001), and symptoms related to XBSMA seem to et al. 1999; Hsing et al. 2000a; Beilin et al. 2001; Latil
start at 38 to 40 CAG repeats (Pioro et al. 1994). et al. 2001; Balic et al. 2002). Seven of these described
Within the normal range of the AR polyglutamine an independent contribution of the CAG repeat poly-
stretch, significant differences between ethnic groups morphism to prostate cancer: either to the age of
have been observed. For healthy men of African des- onset or to the general risk of development. The age
cent the mean number of CAG repeats ranges of the study group, time point and intensity of diag-
between 18 and 20 (Edwards et al. 1992; Platz et al. nostic performance varying with the location of the
2000) and seems to be even shorter in certain African study most likely influence the result as to whether it

43
Chapter 3: Pathophysiology of the androgen receptor

is seen as earlier onset or higher risk. The putative activity represents an important cofactor that takes
association with disease stage is also likely to be influ- positive effect on the supporting function of Sertoli
enced by such factors. Each triplet may hence account cells. Thus, it can be speculated that the CAG repeat
for a 3 to 14% risk for prostate cancer (Stanford et al. polymorphism within the AR gene could have a
1997). In conclusion, it is likely that the genesis of limited influence on spermatogenesis. Such an effect
prostate cancer cells is not induced by androgens, but can be observed as severely impaired spermatogenesis
that stronger androgenicity induced by ARs with in XSBMA patients (Arbizu et al. 1983). The investi-
shorter polyglutamine stretches contributes to a faster gation of the possible influence of a polyglutamine
development of these cells, and this might be seen stretch of within the normal length on sperm produc-
either as earlier onset of or as higher risk for prostate tion requires a sample of carefully selected patients
cancer, depending on the age of the study group. in which significant confounders (obstructive symp-
Epidemiological findings in research on the inci- toms due to infections, congenital aplasia of the vas
dence of prostate cancer in humans confirm an influ- deferens (CBAVD), impaired spermatogenesis due to
ence of the AR gene (CAG)n polymorphism: a meta- hormone disorders, deletions in one of the azoosper-
analysis described an odds ratio of 1.19 for prostate mia-associated regions of the Y chromosome) have
cancer with decreasing (CAG)n (Zeegers et al. 2004). been ruled out.
The modulatory effect on androgen-dependent gene Control groups consisting of healthy fertile males
transcription is described to be linear and obviously should be homogenous in terms of ethnic origin (see
mediated by a differential affinity of coactivator pro- above). It should be considered that within the cohort
teins to the polyglutamine stretch (e.g. ARA24 and of fertile controls, sperm densities below 20 million/
p160) (Hsiao et al. 1999; Irvine et al. 2000). ml might occur (Rajpert-De Meyts et al. 2002).
Another aspect is the putative relation between Unfortunately, a fraction of studies on this subject
BPH and the CAG repeat polymorphism of the AR did not exclude the above-described patients strictly
gene. BPH consists of the overgrowth of tissue within enough. Therefore, it is not surprising that conflicting
the transition zone and periurethral area of the pro- results emerged when infertile and fertile men were
state. This is histologically defined as epithelial and compared in regard to their number of CAG repeats.
fibromuscular hyperplasia (Price et al. 1990). One Some studies reported higher numbers of CAG trip-
factor modulating androgenic exposure is the cellular lets in infertile men (Tut et al. 1997; Legius et al. 1999;
level of androgens, particularly DHT. The influence of Dowsing et al. 1999; Yoshida et al. 1999; Yong et al.
the CAG repeat polymorphism causes variations in 2000; Mifsud et al. 2001; Patrizio et al. 2001; Waller-
such effects as demonstrated by several studies. The and et al. 2001; Mengual et al. 2003), but some did not
two largest studies comparing matched healthy con- (Lundberg-Giwercman et al. 1998; Dadze et al. 2000;
trols (n = 1041 and n = 499) and BPH patients (n = 310 Sasagawa et al. 2001; van Golde et al. 2002; Rajpert-
and n = 449) described the odds ratio for BPH surgery De Meyts et al. 2002). A more recent meta-analysis
or an enlarged prostate gland to be 1.92 (p = 0.0002) involving 33 reports provides support for an associ-
when comparing CAG repeat length of 19 or less to 25 ation between increased AR gene CAG length and
or more. For a six-repeat decrement in CAG repeat idiopathic male infertility; suggesting that even subtle
length, the odds ratio for moderate or severe urinary disruptions in the androgen axis may compromise
obstructive symptoms from an enlarged prostate gland male fertility (Davis-Dao et al. 2007).
was 3.62 (p = 0.004) (Giovannucci et al. 1999a;1999b). When only fertile men covering the whole range
Similarly, adenoma size was found to be inversely asso- of normal sperm concentrations were involved in
ciated with the number of CAG repeats in 176 patients evaluations, a shorter CAG repeat tract was associated
vs. 41 controls (Mitsumori et al. 1999). Prostate growth with higher sperm numbers (von Eckardstein et al.
during androgen substitution is also significantly modi- 2001; Rajpert-De Meyts et al. 2002). Nevertheless,
fied by the CAG repeat polymorphism (see below). a marked variation of sperm density in relation to
the AR polymorphism was observed. Hence, sperm-
atogenesis is likely to be influenced by the number of
3.5.5 Reproductive functions CAG repeats within the normal range, but whether
Stimulation of Sertoli cells by FSH is a prerequisite in this reaches relevance for individuals remains doubt-
primate spermatogenesis, and intratesticular androgen ful. The range of sperm concentrations leading to

44
Chapter 3: Pathophysiology of the androgen receptor

infertility is most likely reached at CAG repeat peak bone density in males will be, while it is incon-
numbers that are associated with XSBMA. Further- clusive whether this effect reaches clinical significance
more, it can be assumed that the proportion of men in terms of higher fracture risk.
with longer CAG repeats among infertile patients It has been demonstrated in a larger cohort of
may, in the case of strict selection criteria excluding soldiers that, even in healthy younger men, shorter
all known causes of infertility, appear higher than in a AR gene CAG repeats may be preventive of stress
control population. Genetic counseling concerning fractures: Smaller-sized (< 16) AR CAG repeats were
inheritability of this modulator of spermatogenesis more prevalent among control subjects (23%) than
is of very restricted value, since the CAG polymorph- among patients (13%); the risk for having stress frac-
ism is located on the X chromosome and the specific ture was almost halved if the size of the repeat was
tract length will affect spermatogenesis of the off- shorter than 16 repeats (Yanovich et al. 2011).
spring only as early as in one half of the grandsons. In women, low androgen levels and, hence, low
activation of the AR are present. In addition, two
alleles of the AR gene will cause an effect less clear
3.5.6 Bone tissue in terms of influence exerted by the CAG repeat
Polymorphisms of the estrogen receptor (ER) have polymorphism. Nevertheless, reports concerning such
repeatedly been demonstrated to modulate quantity impact on bone density in women exist (Sowers et al.
and quality of bone tissue in healthy men (e.g. Sapir- 1999; Chen et al. 2003). As can be expected from
Koren et al. 2001). As androgen activity influences physiology, the AR polymorphism does not influence
bone metabolism (see Chapter 8), these observations the effects of hormone replacement therapy by
apply to the CAG repeat polymorphism in the AR estrogens on bone tissue in postmenopausal women
gene as well: in 110 healthy younger males, a high (Salmen et al. 2003).
number of CAG repeats was significantly associated In addition, the CAG repeat polymorphism affects
with lower bone density (Zitzmann et al. 2001a). This body composition in young men: absolute muscle
result is corroborated by a negative association (thigh) and (lower) trunk increase as CAG(n)
between AR CAG repeat length and bone density at decreases. Expressed relatively, muscle areas and lean
the femoral neck in a group of 508 Caucasian men body mass increase, while fat mass decreases as CAG
aged over 65 years (Zmuda et al. 2000a). The same (n) decreases. The polymorphism does not affect
workgroup also observed a more pronounced bone deep adipose tissues or circulating androgen levels
loss at the hip and increased vertebral fracture risk in young men (Nielsen et al. 2010).
among older men with longer AR CAG repeat length
(Zmuda et al. 2000b). In a group of 140 Finnish men
aged 5060 years, lumbar and femoral bone mineral 3.5.7 Cardiovascular risk factors
density values were higher in those men with shorter Testosterone plays an ambiguous role in relation to
CAG repeats in comparison to those with longer cardiovascular risk factors, and its respective role has
CAG repeats (Remes et al. 2003). The differences not been fully resolved (see Chapter 10). The interactions
reach statistical significance when the groups with between the CAG repeat polymorphism, serum levels of
CAG repeat length of 1517 and 2226 are compared sex hormones, lifestyle factors and endothelium-depend-
directly. In contrast, in a group of 273 healthy Belgian ent and independent vessel relaxation of the brachial
men aged 71 and 86 years, no influence of the andro- artery, as well as lipoprotein levels, leptin and insulin
gen receptor gene polymorphism was seen (Van concentrations and body composition were described
Pottelbergh et al. 2001). in over 100 eugonadal men of a homogenous population.
Higher androgenization will lead to higher peak In agreement with previously demonstrated androgen
bone mass (Khosla 2002); thus, the AR polymorphism effects on these parameters, it was demonstrated that
effects on bone density are likely to be visible among androgenic effects were attenuated in persons with
healthy younger males, while the difference could be longer CAG repeats, while testosterone levels themselves
mitigated by the overall age-dependent bone loss and played only a minor role within the eugonadal range.
may be no longer visible in old men, in whom con- A marked increased prevalence of the metabolic
founders have exerted influence on bone tissue. Thus, syndrome in older men was seen in those patients with
the longer the CAG repeat in the AR gene, the lower longer CAG repeats than average (Stanworth et al. 2008).

45
Chapter 3: Pathophysiology of the androgen receptor

Significant positive correlations with the length of be speculated that the higher activation rate of the AR
CAG repeats were seen for endothelial-dependent in this subgroup revealed effects of declining andro-
vasodilatation, HDL-cholesterol concentrations, body gen levels more readily than in subgroups with longer
fat content, insulin and leptin levels. These results CAG repeats.
remained stable in multiple regression analyses In a sample of 172 Finnish men aged 41 to 70
correcting for age and lifestyle factors. It was demon- years, the length of CAG repeats was significantly,
strated by a five-factor model that adverse and bene- and independently of testosterone levels, positively
ficial components are mutually dependent (Zitzmann associated with symptom scores concerning depres-
et al. 2001b; 2003a). Within the investigated range sion, as expressed by the wish to be dead (r = 0.45; p <
of androgen-related cardiovascular risk factors and 0.0001), depressed mood (r = 0.23; p = 0.003), anxiety
eugonadal testosterone levels, the CAG repeat poly- (r = 0.15; p < 0.05), deterioration of general well-
morphism could play a more dominant role than being (r = 0.22; p = 0.004) and also decreased beard
testosterone itself. Concerning lipid concentrations, growth (r = 0.49; p < 0.0001) (Harkonen et al. 2003).
corresponding results are reported in men with This was later confirmed by a large epidemiological
XSBMA (Dejager et al. 2002). Also in agreement, an trial involving patients from both psychological and
inclination to develop diabetes mellitus has been andrological care units in comparison to healthy con-
described for these patients (Arbizu et al. 1983). trols: scores of the Aging Male Symptom Score as well
Hence, adverse or beneficial effects of a longer or as Depression Scales were markedly elevated in sub-
shorter CAG repeat chain in regard to cardiovascular jects with both lower testosterone concentrations and
risk will most likely strongly depend on cofactors. The longer CAG repeats (Schneider et al. 2011a; 2011b).
implications in terms of modulation of cardiovascular Another aspect of psychological parameters is rep-
risk by androgens apply especially to hypogonadal resented by the group of externalizing behaviors; these
men receiving testosterone substitution. The pharma- are predominantly found in males and have been asso-
cogenetic role in this respect of the CAG repeat poly- ciated with androgens (Zitzmann and Nieschlag 2001).
morphism has yet to be elucidated. The personality traits are attention deficit hyperactivity
disorder (ADHD); conduct disorder (CD) and oppos-
itional defiant disorder (ODD). A controlled study in
3.5.8 Psychological implications 302 younger men concerning these disorders in rela-
Testosterone substitution in hypogonadal men tion to the CAG repeat of the AR gene demonstrated
improves lethargic or depressive aspects of mood a significantly higher prevalence in genotypes with
significantly (Burris et al. 1992). Studies exploring shorter repeat chains. The group also reported an
the relationship between gonadal function and association of short CAG repeats in the AR gene with
depressive episodes demonstrated testosterone levels novelty-seeking behavior (drug abuse, pathological
to be markedly decreased in these patients (Unden gambling) (Comings et al. 1999).
et al. 1988; Schweiger et al. 1999; Barrett-Connor et al. Confirmingly, in a sample of 183 healthy Swedish
1999). Accordingly, treatment with testosterone gel men aged 2075 years, associations of CAG repeat
may improve symptoms in men with refractory length and scores in the Karolinska Scales of Person-
depression (Pope et al. 2003). The age-dependent ality were described. Tendencies indicated positive
decline of testosterone levels is sometimes associated relationships between shorter CAG trinucleotide
with symptoms of depression. It has been recently repeats and personality scales connected to dominance
demonstrated in 1000 older men that this mood and aggression (low Lack of Assertiveness; high
dependency on androgen levels is modified by the Verbal Aggression; high Monotony Avoidance).
CAG repeat polymorphism of the AR gene. Depres- Longer polyglutamine tracts were associated with
sion scores were significantly and inversely associated some neuroticism-related personality scales: high
with testosterone levels in subjects with shorter CAG Muscular Tension, high Lack of Assertiveness
repeats, while this was not observed in men with and high Psychasthenia (Jnsson et al. 2001).
moderate and longer polyglutamine stretches in the In addition, there exists a case report of three
AR protein. Low versus high testosterone in such Caucasian brothers with mental retardation especially
men was associated with a fivefold increased likeli- demonstrating a delay in speech development, shy
hood of depressive mood (Seidman et al. 2001). It can but sometimes aggressive behavior, marfanoid

46
Chapter 3: Pathophysiology of the androgen receptor

habitus and relatively large testes in combination marked influence of the CAG repeat polymorphism
with abnormally short CAG repeats (eight triplets) on prostate growth (Zitzmann et al. 2003b).
(Kooy et al. 1999). Another retrospective approach concerning phar-
macogenetic influences in hormonal male contracep-
tion demonstrated, as spermatogenesis is partially
3.5.9 Hair growth dependent on intratesticular androgen activity, sperm
Male pattern baldness is described by a loss of scalp counts to be more easily suppressed by various
hair and affects up to 80% of males by the age of 80 pharmacological regimens in men with longer CAG
years. A balding scalp is caused by androgens and repeats in the subgroup with remnant gonadotropin
expression of the AR in the respective hair follicle, activity (von Eckardstein et al. 2002).
and is thus known as androgenetic alopecia (see Testosterone levels within the normal range will
Chapter 7). One can assume that the influence of more or less saturate the androgen receptors present,
the CAG repeat polymorphism on androgenicity and it has been demonstrated that androgenic effects
causes a variation of androgenetic alopecia. In men will reach a plateau at certain levels, which are prob-
with such a clinical condition, significantly shorter ably tissue specific (Zitzmann et al. 2002a; 2002b). In
CAG repeats were described in comparison to con- agreement with this, a study applying exponentially
trols by two studies (Sawaya and Shalita 1998; Ellis increasing doses of testosterone to hypogonadal men
et al. 2001). Thus, the CAG repeat polymorphism shows corresponding results (Bhasin et al. 2001):
is likely to play a role in modulation of androgen androgen effects on various parameters increased lin-
influence on male hair pattern, but since statistical early with the logarithm of testosterone levels and
significance is weak in a reasonable number of patients linearly with the logarithm of the testosterone dose.
due to high interindividual variability, the cosmetic In practice, this means more or less a plateau effect.
consequence for the individual is questionable. Significant increments of androgenic effects caused by
rising testosterone levels within the eugonadal range
are seen only when the normal range is left and clearly
3.5.10 Pharmacogenetics and hypotheses supraphysiological levels are reached. Therefore, it
Considering the observations in eugonadal men, one can be assumed that within the range of such a plat-
can assume that testosterone therapy in hypogonadal eau of saturation, genetically determined functional
men should have a differential impact on androgen differences in androgen receptor activity can be best
target tissue, depending on the number of CAG observed; while in a condition of hypogonadism,
repeats. In a longitudinal pharmacogenetic study in androgenicity will be strongly dependent on androgen
131 hypogonadal men, prostate volume was assessed levels themselves, as binding to and, hence, activation
before and under androgen substitution. The length of androgen receptors will increase until saturation is
of CAG repeats, sex hormone levels and anthropo- reached (Fig. 3.3). This model explains why androgen
metric measures were considered. Initial prostate size effects are found between hypo- and eugonadal men
of hypogonadal men was dependent on age and base- but can often not be confirmed for various testoster-
line testosterone levels, but not the CAG repeat poly- one levels within the eugonadal range. During substi-
morphism. However, when prostate size increased tution therapy of hypogonadal men, both effects on
significantly during therapy, prostate growth per year androgenicity increment of testosterone levels from
and absolute prostate size under substituted testoster- the hypo-into the eugonadal range and modulation of
one levels were strongly dependent on the AR poly- androgen effects within the eugonadal range by the
morphism, with lower treatment effects in patients androgen receptor polymorphism have to be taken
with longer repeats. Other modulators of prostate into account.
growth were age and testosterone level under treat- Studies examining the effects of the AR gene
ment. The odds ratio for men with repeats < 20, (CAG)n polymorphism have to consider androgen
compared to those with  20, to develop a prostate levels for a proper description of clinical relevance,
size of at least 30 ml under testosterone substitution because involving hypogonadal men will distort
was 8.7 (95% confidence interval (CI) 3.124.3; p < results owing to lack of sufficient ligand binding,
0.001). This first pharmacogenetic study on androgen hence, activation of the AR. Similarly, not considering
substitution in hypogonadal men demonstrates a testosterone levels, possibly compensating for the

47
Chapter 3: Pathophysiology of the androgen receptor

Short CAG repeats lead to enhanced effects in estrogen-dependent tissues,


such as bones (Limer et al. 2009).
However, investigation of men with an intact HPG
axis does not focus on the clinically relevant clientele:
those men with symptoms of androgen deficiency,
Androgen effect

those men with disturbances of the HPG axis. Such


Long CAG repeats
disorders can be of a milder nature, as seen in late-onset
hypogonadism (LOH) (Wang et al. 2008) or subjects
with the metabolic syndrome (Shabsigh et al. 2008),
conditions in which both pituitary function and Leydig
cell capacity are impaired. In case of longer (CAG)n,
these men will present with inadequately low-normal
LH concentrations and/or low-normal testosterone
levels, but, nevertheless, will require higher concentra-
Hypo- Eugonadal range tions of testosterone to compensate for their attenuated
Testosterone levels androgen action. Thus, they are likely to present with
Fig. 3.3 Hypothetical model of androgen effects: within the features of hypogonadism in the presence of normal
hypogonadal range, and in comparison to the eugonadal range, testosterone levels (Canale et al. 2005). Exactly this is
differences in androgen effects are determined by testosterone the patient group most likely missed for investigation,
levels. Within the eugonadal range, androgen effects depend
rather on the AR polymorphism. As this effect depends on the diagnostics and putative treatment up to this day.
presence of AR coactivators, the concentrations of which are In the case of classical hypogonadism (i.e. the
tissue specific, the shapes of the curves are putatively variable from (almost) complete breakdown of the HPG axis due to
organ to organ.
causes of primary or secondary origin), testosterone
levels are low and patients require substitution. The
effects of the (CAG)n polymorphism, takes an incom- findings in men with intact HPG axes demonstrate
plete approach. When examining the effects of andro- that men with longer (CAG)n require higher testoster-
gens, regression models are required to include both one concentrations for normal androgen action in
testosterone concentrations and the length of the AR comparison to men with shorter (CAG)n (Huhta-
gene CAG repeat polymorphism. In this light, a recent niemi et al. 2009). Thus, hypogonadal persons with
study in a large cohort of aging men did not demon- longer (CAG)n will then, as do their healthy counter-
strate a relationship of the AR gene CAG repeat poly- parts, need higher testosterone levels (and, hence,
morphism to the risk of heart disease, possibly due to higher testosterone doses) to compensate for mitigated
the fact that serum testosterone concentrations were androgen action. Correspondingly, persons with
not considered (Page et al. 2006). rather short (CAG)n will need lower doses of testos-
The weaker androgen action induced by longer terone substitution in the case of hypogonadism.
(CAG)n will also affect the feedback mechanism of
the hypothalamic-pituitary-gonadal (HPG) axis. In
healthy men, longer (CAG)n will usually provoke 3.6 Outlook
higher LH secretion (Zitzmann et al. 2001b; Stan- Further decoding of the molecular and biochemical
worth et al. 2008; Huhtaniemi et al. 2009). This pathways is necessary for a comprehensive under-
can, in persons with intact and fully responsive standing of normal and abnormal sexual determin-
Leydig cell capacity, result in higher concentra- ation and differentiation. Based on the known
tions of testosterone and, hence, compensation of molecular defects of impaired human sexual develop-
weaker androgen action. As a result of higher ment, recent achievements in the field of functional
testosterone concentrations, its aromatization genomics and proteomics offer unique opportunities
product estradiol will also be present in higher to identify the genetic programs downstream of these
amounts (Huhtaniemi et al. 2009). Such higher pathways, which are ultimately responsible for struc-
estradiol concentrations in these men with intact ture and function of a normal or abnormal genital
HPG regulation or feedback mechanisms can even phenotype. Hopefully this knowledge will lead to better

48
Chapter 3: Pathophysiology of the androgen receptor

Increasing number of CAG triplets XSBMA Fig. 3.4 The inverse association
between the number of CAG repeats in
the AR gene and functionality of the AR
protein. Longer CAG tracts result in lower
Normal range
38 transcription of target genes and, thus,
9 37 lower androgenicity. Expansion of the
encoded polyglutamine stretch to
beyond probably 38 leads to the
neuromuscular disorder X-linked spinal
Decreasing androgenicity and bulbar muscular atrophy (XSBMA); a
condition in which defective
spermatogenesis and undervirilization are
Affected parameters: Neurological observed. Conversely, low numbers of
Prostate growth and prostate cancer disorders CAG repeats are associated with
increased androgenicity of susceptible
Spermatogenesis Gynecomastia tissues.
Bone tissue
Diabetes
Lipid metabolism and body composition mellitus
Vascular endothelial functions
Personality traits Ineffective
spermatogenesis
Hair growth

medical decisions in patients with androgen insensitiv-


ity due to AR defects, and will open pathways for the  During childhood, laboratory analysis is often
difficult and lacks specificity and sensitivity.
development of individual therapeutic options.
 In adolescence and adulthood, diagnosis
The highly polymorphic nature of glutamine resi-
of androgen insensitivity can be made
dues within the AR protein, which is encoded by the on the grounds of undermasculinization
CAG repeat polymorphism within the AR gene, causes and/or feminization despite high measurable
a subtle gradation of androgenicity among individuals. androgen levels.
This modulation of androgen effects may be small but  Molecular genetic analysis of the androgen
continuously present during a mans lifetime, thus receptor gene can detect a relevant mutation
exerting effects that are measurable in many tissues only in a subset of patients. The less severe the
as various degrees of androgenicity (Fig. 3.4). It phenotype, the less likely is the chance of
remains to be seen whether these insights are import- demonstrating a mutation.
ant enough to become part of individually useful  Highly specific tools for early and specific
detection of androgen receptor abnormalities
laboratory assessments. The pharmacogenetic impli-
must be developed.
cation of this polymorphism seems to play an import-
 High-throughput analysis is needed to elucidate
ant role as modulator of treatment effects in genetic alterations in factors leading to androgen
hypogonadal men. Further studies are required to insensitivity without mutation of the androgen
decide whether these insights should sublimate into receptor.
individualized aspects of testosterone therapy; for  Future research should evolve model systems
example adaptation of dosage or surveillance intervals. to better elucidate individualized
genotypephenotype correlations.
3.7 Key messages  The characterization of other factors of
 Androgen insensitivity has a very variable androgen-controlled genital development is
phenotypic appearance and may be mandatory for further insight into the cellular and
clinically undistinguishable from time-dependent events of androgen action and
defects of androgen biosynthesis during to define further mechanisms of androgen
childhood. insensitivity.

49
Chapter 3: Pathophysiology of the androgen receptor

 The CAG repeat polymorphism in exon 1 of the  The pharmacogenetic implications of this
AR gene modulates androgen effects: longer polymorphism are likely to play a significant role in
triplets attenuate testosterone effects. future testosterone treatment of hypogonadal men.

3.8 References putative biomarker of androgen


receptor function in androgen
carcinoma subjects. Cancer
92:941949
Abdullah A, Trifiro MA, Panet- insensitivity syndrome. J Mol Med
Raymond V, Alvarado C, de Bhasin S, Woodhouse L, Casaburi R,
87:623632 Singh AB, Bhasin D, Berman N,
Tourreil S, Frankel D, Schipper
HM, Pinsky L (1998) Spinobulbar Arbizu T, Santamaria J, Gomez JM, Chen X, Yarasheski KE, Magliano L,
muscular atrophy: polyglutamine- Quilez A, Serra JP (1983) A family Dzekov C, Dzekov J, Bross R,
expanded androgen receptor is with adult onset spinal and bulbar Phillips J, Sinha-Hikim I, Shen R,
proteolytically resistant in vitro and muscular atrophy X-linked Storer TW (2001) Testosterone dose-
processed abnormally in transfected inheritance and associated testicular response relationships in healthy
cells. Hum Mol Genet 7:379384 failure. J Neurol Sci 59:371382 young men. Am J Physiol Endocrinol
Balic I, Graham ST, Troyer DA, Metab 281:11721181
Adachi M, Takayanagi R, Tomura A,
Imasaki K, Kato S, Goto K, Yanase Higgins BA, Pollock BH, Johnson- Bhatia-Gaur R, Donjacour AA,
T, Ikuyama S, Nawata H (2000) Pais TL, Thompson IM, Leach RJ Sciavolino PJ, Kim M, Desai N,
Androgen-insensitivity syndrome as (2002) Androgen receptor length Young P, Norton CR, Gridley T,
a possible coactivator disease. polymorphism associated with Cardiff RD, Cunha GR, Abate-Shen
N Engl J Med 343:856862 prostate cancer risk in Hispanic C, Shen MM (1999) Roles for
men. J Urol 168:22452248 Nkx3.1 in prostate development and
Ahmed SF, Achermann JC, Arlt W, cancer. Genes Dev 13:966977
Balen A, Conway G, Edwards Z, Barrett-Connor E, von Mhlen DG,
Elford S, Hughes IA, Izatt L, Krone Kritz-Silverstein D (1999) Bieberich CJ, Fujita K, He WW, Jay G
N, Miles H, OToole S, Perry L, Bioavailable testosterone and (1996) Prostate-specific and
Sanders C, Simmonds M, Michael depressed mood in older men: the androgen-dependent expression of a
Wallace A, Watt A, Willis D (2011) Rancho Bernardo Study. J Clin novel homeobox gene. J Biol Chem
UK guidance on the initial Endocr Metab 84:573577 271:3177931782
evaluation of an infant or an Barsoum IB, Yao HH (2010) Fetal Boehmer AL, Brinkmann O,
adolescent with a suspected disorder Leydig cells: progenitor cell Bruggenwirth H, van Assendelft C,
of sex development. Clin Endocrinol maintenance and differentiation. Otten BJ, Verleun-Mooijman MC,
(Oxf) 75:1226 J Androl 31:1115 Niermeijer MF, Brunner HG,
Aiman J, Griffin JE, Gazak JM, Wilson Barsoum IB, Bingham NC, Parker KL, Rouwe CW, Waelkens JJ, Oostdijk W,
JD, MacDonald PC (1979) Jorgensen JS, Yao HH (2009) Kleijer WJ, van der Kwast TH, de
Androgen insensitivity as a cause of Activation of the Hedgehog Vroede MA, Drop SL (2001)
infertility in otherwise normal men. pathway in the mouse fetal ovary Genotype versus phenotype in
N Engl J Med 300:223227 leads to ectopic appearance of fetal families with androgen insensitivity
Leydig cells and female syndrome. J Clin Endocrinol Metab
Albertelli MA, Scheller A, Brogley M,
pseudohermaphroditism. Dev Biol 86:41514160
Robins DM (2006) Replacing the
mouse androgen receptor with 329:96103 Boukari K, Meduri G, Brailly-Tabard S,
human alleles demonstrates Bebermeier JH, Brooks JD, Deprimo Guibourdenche J, Ciampi ML,
glutamine tract length-dependent SE, Werner R, Deppe U, Demeter J, Massin N, Martinerie L, Picard JY,
effects on physiology and Hiort O, Holterhus PM (2006) Cell- Rey R, Lombes M, Young J (2009)
tumorigenesis in mice. Mol line and tissue-specific signatures of Lack of androgen receptor expression
Endocrinol 20:12481260 androgen receptor-coregulator in Sertoli cells accounts for the
Andersson S, Berman DM, Jenkins EP, transcription. J Mol Med absence of anti-Mullerian hormone
Russell DW (1991) Deletion of 84:919931 repression during early human testis
steroid 5 alpha-reductase 2 gene in development. J Clin Endocrinol
Beilin J, Harewood L, Frydenberg M,
male pseudohermaphroditism. Metab 94:18181825
Mameghan H, Martyres RF, Farish
Nature 354:159161 SJ, Yue C, Deam DR, Byron KA, Bouvattier C, Carel JC, Lecointre C,
Appari M, Werner R, Wunsch L, Zajac JD (2001) A case-control David A, Sultan C, Bertrand AM,
Cario G, Demeter J, Hiort O, Riepe study of the androgen receptor Morel Y, Chaussain JL (2002)
FJ, Holterhus PM (2009) gene CAG repeat polymorphism Postnatal changes of T, LH, and
Apolipoprotein D (APOD) is a in Australian prostate FSH in 46,XY infants with

50
Chapter 3: Pathophysiology of the androgen receptor

mutations in the AR gene. J Clin regulation. J Steroid Biochem Mol gene shows no significant
Endocrinol Metab 87:2932 Biol 76:2330 relationship to impaired
Bullejos M, Koopman P (2001) Spatially Claessens F, Denayer S, Van Tilborgh spermatogenesis in an infertile
dynamic expression of Sry in mouse N, Kerkhofs S, Helsen C, Haelens A Caucasoid sample of German
genital ridges. Dev Dyn 221:201205 (2008) Diverse roles of androgen origin. Mol Hum Reprod 6:207214

Burris AS, Banks SM, Carter CS, Davidson receptor (AR) domains in AR- Davis-Dao CA, Tuazon ED, Sokol RZ,
JM, Sherins RJ (1992) A long-term mediated signaling. Nucl Recept Cortessis VK (2007) Male infertility
prospective study of the physiologic Signal 6:e008 and variation in CAG repeat length
and behavioural effects of hormone Comings DE, Chen C, Wu S, in the androgen receptor gene: a
replacement in untreated hypogonadal Muhleman D (1999) Association of meta-analysis. J Clin Endocrinol
men. J Androl 13:297304 the androgen receptor gene (AR) Metab 92:43194326
Canale D, Caglieresi C, Moschini C, with ADHD and conduct disorder. Dejager S, Bry-Gauillard H, Bruckert E,
Liberati CD, Macchia E, Pinchera A, Neuroreport 10:15891592 Eymard B, Salachas F, LeGuern E,
Martino E (2005) Androgen Cools M, Wolffenbuttel KP, Drop SL, Tardieu S, Chadarevian R, Giral P,
receptor polymorphism (CAG Oosterhuis JW, Looijenga LH Turpin G (2002) A comprehensive
repeats) and androgenicity. Clin (2011) Gonadal development and endocrine description of Kennedys
Endocrinol (Oxf) 63:356361 tumor formation at the crossroads disease revealing androgen
of male and female sex insensitivity linked to CAG repeat
Chamberlain NL, Driver ED, Miesfeld RL length. J Clin Endocr Metab
(1994) The length and location of determination. Sex Dev 5:167180
87:38933901
CAG trinucleotide repeats in the Correa-Cerro L, Wohr G, Haussler J,
androgen receptor N-terminal domain Berthon P, Drelon E, Mangin P, Deslypere JP, Young M, Wilson JD,
affect transactivation function. Nucleic Fournier G, Cussenot O, Kraus P, McPhaul MJ (1992) Testosterone
Acids Res 22:31813186 Just W, Paiss T, Cantu JM, Vogel W and 5 alpha-dihydrotestosterone
(1999) (CAG)nCAA and GGN interact differently with the
Chamberlain NL, Whitacre DC,
repeats in the human androgen androgen receptor to enhance
Miesfeld RL (1996) Delineation of
receptor gene are not associated transcription of the MMTV-CAT
two distinct type 1 activation
with prostate cancer in a French- reporter gene. Mol Cell Endocrinol
functions in the androgen receptor
German population. Eur J Hum Gen 88:1522
amino-terminal domain. J Biol
Chem 271:2677226778 7:357362 Doehnert U, Bertelloni S, Richter-
Coutinho-Camillo CM, Miracca EC, Unruh A, Werner R, Hiort O (2011)
Chang CS, Kokontis J, Liao ST (1988)
dos Santos ML, Salaorni S, Sarkis Hormone profiles in adolescents
Molecular cloning of human and rat
AS, Nagai MA (2006) Identification and adults with complete androgen
complementary DNA encoding
of differentially expressed genes in insensitivity syndrome. In: Hiort O
androgen receptors. Science
prostatic epithelium in relation to (ed) 3rd International Symposium
240:324326
androgen receptor CAG repeat on Disorders of Sex Development,
Chen HY, Chen WC, Wu MC, Tsai FJ, length. Int J Biol Markers 21:96105 Lbeck. Book of abstracts,
Tsai CH (2003) Androgen receptor Universitt zu Lbeck, Lbeck,
(AR) gene microsatellite Cutress ML, Whitaker HC, Mills IG,
Germany, p 57
polymorphism in postmenopausal Stewart M, Neal DE (2008)
Structural basis for the nuclear Dowsing AT, Yong EL, Clark M,
women: correlation to bone mineral
import of the human androgen McLachlan RI, de Kretser DM,
density and susceptibility to
osteoporosis. Eur J Obstet Gynecol receptor. J Cell Sci 121:957968 Trounson AO (1999) Linkage
between male infertility and
Reprod Biol 107:5256 da Costa Rose Paulino M, Steinmetz L,
trinucleotide repeat expansion in
Choong CS, Wilson EM (1998) Menezes Filho HC, Kuperman H,
the androgen-receptor gene. Lancet
Della Manna T, Vieira JG,
Trinucleotide repeats in the human 354:640643
androgen receptor: a molecular Blasbalg R, Baroni R, Setian N,
basis for disease. J Mol Endocrinol Damiani D (2009) Search of Doyu M, Sobue G, Mukai E, Kachi T,
prostatic tissue in 46,XX congenital Yasuda T, Mitsuma T, Takahashi A
21:235257
adrenal hyperplasia. Arq Bras (1992) Severity of X-linked recessive
Claessens F, Verrijdt G, Schoenmakers Endocrinol Metabol 53:716720 bulbospinal neuronopathy
E, Haelens A, Peeters B, Verhoeven correlates with size of the tandem
G, Rombauts W (2001) Selective Dadze S, Wieland C, Jakubiczka S,
CAG repeat in androgen receptor
DNA binding by the Funke K, Schrder E, Royer-
Pokora B, Willers R, Wieacker PF gene. Annals Neurol 32:707710
androgen receptor as a mechanism
for hormone-specific gene (2000) The size of the CAG repeat Edwards A, Hammond HA, Jin L,
in exon 1 of the androgen receptor Caskey CT, Chakraborty R (1992)

51
Chapter 3: Pathophysiology of the androgen receptor

Genetic variation at five trimeric Coleman N, Hughes IA (2006) Inherited and de novo androgen
and tetrameric tandem repeat loci in Testicular development in the receptor gene mutations:
four human population groups. complete androgen insensitivity investigation of single-case families.
Genomics 12:241253 syndrome. J Pathol 208:518527 J Pediatr 132:939943
Ellis JA, Stebbing M, Harrap SB (2001) Haraguchi R, Mo R, Hui C, Hiort O, Holterhus PM, Horter T,
Polymorphism of the androgen Motoyama J, Makino S, Shiroishi T, Schulze W, Kremke B, Bals-
receptor gene is associated with Gaffield W, Yamada G (2001) Pratsch M, Sinnecker GH, Kruse K
male pattern baldness. J Invest Unique functions of Sonic (2000) Significance of mutations in
Dermatol 116:452455 hedgehog signaling during external the androgen receptor gene in males
Giovannucci E, Stampfer MJ, genitalia development. Development with idiopathic infertility. J Clin
Krithivas K, Brown M, Dahl D, 128:42414250 Endocrinol Metab 85:28102815
Brufsky A, Talcott J, Hennekens CH, Harkonen K, Huhtaniemi I, Makinen J, Hiort O, Reinecke S, Thyen U,
Kantoff PW (1997) The CAG repeat Hbler D, Irjala K, Koskenvuo M, Jurgensen M, Holterhus PM,
within the androgen receptor gene Oettel M, Raitakari O, Saad F, Schon D, Richter-Appelt H (2003)
and its relationship to prostate cancer. Pollanen P (2003) The polymorphic Puberty in disorders of somatosexual
Proc Natl Acad Sci 94:33203323 androgen receptor gene CAG repeat differentiation. J Pediatr Endocrinol
Giovannucci E, Stampfer MJ, Chan A, pituitary-testicular function and Metab 16 (Suppl 2):297306
Krithivas K, Gann PH, Hennekens andropausal symptoms in ageing Holterhus PM, Wiebel J, Sinnecker
CH, Kantoff PW (1999a) CAG men. Int J Androl 26:187194 GH, Bruggenwirth HT, Sippell WG,
repeat within the androgen receptor He B, Gampe RT Jr, Kole AJ, Hnat AT, Brinkmann AO, Kruse K, Hiort O
gene and incidence of surgery for Stanley TB, An G, Stewart EL, (1999) Clinical and molecular
benign prostatic hyperplasia in U.S. Kalman RI, Minges JT, Wilson EM spectrum of somatic mosaicism in
physicians. Prostate 39:130134 (2004) Structural basis for androgen androgen insensitivity syndrome.
Giovannucci E, Platz EA, Stampfer MJ, receptor interdomain and Pediatr Res 46:684690
Chan A, Krithivas K, Kawachi I, coactivator interactions suggests a Holterhus PM, Sinnecker GH, Hiort O
Willett WC, Kantoff PW (1999b) transition in nuclear receptor (2000) Phenotypic diversity and
The CAG repeat within the activation function dominance. Mol testosterone-induced normalization
androgen receptor gene and benign Cell 16:425438 of mutant L712F androgen receptor
prostatic hyperplasia. Urology Hellwinkel OJ, Bull K, Holterhus PM, function in a kindred with androgen
53:121125 Homburg N, Struve D, Hiort O insensitivity. J Clin Endocrinol
Habert R, Lejeune H, Saez JM (2001) (1999) Complete androgen Metab 85:32453250
Origin, differentiation and insensitivity caused by a splice Holterhus PM, Bruggenwirth HT,
regulation of fetal and adult Leydig donor site mutation in intron 2 of Brinkmann AO, Hiort O (2001)
cells. Mol Cell Endocrinol 179:4774 the human androgen receptor gene Post-zygotic mutations and somatic
resulting in an exon 2-lacking mosaicism in androgen insensitivity
Hacker A, Capel B, Goodfellow P, transcript with premature stop- syndrome. Trends Genet 17:627628
Lovell-Badge R (1995) Expression of codon and reduced expression.
Sry, the mouse sex determining J Steroid Biochem Mol Biol 68:19 Holterhus PM, Hiort O, Demeter J,
gene. Development 121:16031614 Brown PO, Brooks JD (2003)
Hellwinkel OJ, Holterhus PM, Differential gene-expression
Haelens A, Verrijdt G, Callewaert L, Struve D, Marschke C, Homburg N, patterns in genital fibroblasts of
Christiaens V, Schauwaers K, Hiort O (2001) A unique exonic normal males and 46,XY females
Peeters B, Rombauts W, Claessens F splicing mutation in the human with androgen insensitivity
(2003) DNA recognition by the androgen receptor gene indicates a syndrome: evidence for early
androgen receptor: evidence for an physiologic relevance of regular programming involving the
alternative DNA-dependent androgen receptor transcript androgen receptor. Genome
dimerization, and an active role of variants. J Clin Endocrinol Metab Biol 4:R37
sequences flanking the response 86:25692575
element on transactivation. Biochem Holterhus PM, Deppe U, Werner R,
Hiort O, Sinnecker GH, Holterhus PM,
J 369:141151 Richter-Unruh A, Bebermeier JH,
Nitsche EM, Kruse K (1996) The Wunsch L, Krege S, Schweikert HU,
Hannema SE, Hughes IA (2007) clinical and molecular spectrum of Demeter J, Riepe F, Hiort O, Brooks
Regulation of Wolffian duct androgen insensitivity syndromes. JD (2007) Intrinsic androgen-
development. Horm Res 67:142151 Am J Med Genet 63:218222 dependent gene expression patterns
Hannema SE, Scott IS, Rajpert-De Hiort O, Sinnecker GH, Holterhus PM, revealed by comparison of genital
Meyts E, Skakkebaek NE, Nitsche EM, Kruse K (1998) fibroblasts from normal males and

52
Chapter 3: Pathophysiology of the androgen receptor

individuals with complete and Imperato-McGinley J, Sanchez RS, bulbar muscular atrophy of late
partial androgen insensitivity Spencer JR, Yee B, Vaughan ED onset. Neurology 18:671680
syndrome. BMC Genomics 8:376 (1992) Comparison of the effects of Khosla S (2002) Oestrogen bones and
Holterhus PM, Bebermeier JH, Werner the 5 alpha-reductase inhibitor men: when testosterone just isnt
R, Demeter J, Richter-Unruh A, finasteride and the antiandrogen enough. Clin Endocrinol 56:291293
Cario G, Appari M, Siebert R, Riepe flutamide on prostate and genital
differentiation: dose-response Kim Y, Kobayashi A, Sekido R,
F, Brooks JD, Hiort O (2009)
studies. Endocrinology DiNapoli L, Brennan J, Chaboissier
Disorders of sex development
expose transcriptional autonomy of 131:11491156 MC, Poulat F, Behringer RR, Lovell-
Badge R, Capel B (2006) Fgf9 and
genetic sex and androgen- Irvine RA, Ma H, Yu MC, Ross RK,
Wnt4 act as antagonistic signals to
programmed hormonal sex in Stallcup MR, Coetzee GA (2000)
regulate mammalian sex
human blood leukocytes. BMC Inhibition of p160-mediated
determination. PLoS Biol 4:e187
Genomics 10:292 coactivation with increasing
androgen receptor polyglutamine Kim Y, Bingham N, Sekido R, Parker
Hsiao PW, Lin DL, Nakao R, Chang C
(1999) The linkage of Kennedys length. Hum Mol Genet 9:267274 KL, Lovell-Badge R, Capel B (2007)
Fibroblast growth factor receptor 2
neuron disease to ARA24, the first Jenster G, van der Korput JA, Trapman
regulates proliferation and Sertoli
identified androgen receptor J, Brinkmann AO (1992) Functional
differentiation during male sex
polyglutamine region-associated domains of the human androgen
determination. Proc Natl Acad Sci
coactivator. J Biol Chem receptor. J Steroid Biochem Mol Biol
USA 104:1655816563
274:2022920234 41:671675
Kittles RA, Young D, Weinrich S,
Hsing AW, Gao YT, Wu G, Wang X, Jenster G, Trapman J, Brinkmann AO
Hudson J, Argyropoulos G, Ukoli F,
Deng J, Chen YL, Sesterhenn IA, (1993) Nuclear import of the
Adams-Campbell L, Dunston GM
Mostofi FK, Benichou J, Chang C human androgen receptor. Biochem
(2001) Extent of linkage
(2000a) Polymorphic CAG and J 293:761768
disequilibrium between the
GGN repeat lengths in the androgen Jenster G, van der Korput HA, androgen receptor gene CAG and
receptor gene and prostate cancer Trapman J, Brinkmann AO (1995) GGC repeats in human populations:
risk: a population-based case- Identification of two transcription implications for prostate cancer
control study in China. Cancer Res activation units in the N-terminal risk. Hum Gen 109:253261
60:51115116 domain of the human androgen
Hsing AW, Tsao L, Devesa SS (2000b) receptor. J Biol Chem Klessen C, Asbach P, Hein PA,
International trends and patterns of 270:73417346 Beyersdorff D, Hamm B, Taupitz M
prostate cancer incidence and (2005) Complex genital
Jnsson EG, von Gertten C, malformation in a female with
mortality. Int J Cancer 85:6067 Gustavsson JP, Yuan QP, Lindblad- congenital adrenal hyperplasia:
Huang L, Pu Y, Hepps D, Danielpour Toh K, Forslund K, Rylander G, evaluation with magnetic resonance
D, Prins GS (2007) Posterior Hox Mattila-Evenden M, Asberg M, imaging. Acta Radiol 46:891894
gene expression and differential Schalling M (2001) Androgen
androgen regulation in the receptor trinucleotide repeat Kohler B, Lumbroso S, Leger J, Audran
developing and adult rat prostate polymorphism and personality F, Grau ES, Kurtz F, Pinto G,
lobes. Endocrinology 148:12351245 traits. Psychiatr Genet 11:1923 Salerno M, Semitcheva T,
Czernichow P, Sultan C (2005)
Huhtaniemi IT, Pye SR, Limer KL, Kashimada K, Koopman P (2010) Sry: Androgen insensitivity syndrome:
Thomson W, ONeill TW, Platt H, the master switch in mammalian sex somatic mosaicism of the androgen
Payne D, John SL, Jiang M, Boonen determination. Development receptor in seven families and
S, Borghs H, Vanderschueren D, 137:39213930 consequences for sex assignment
Adams JE, Ward KA, Bartfai G, Kazemi-Esfarjani P, Trifiro MA, and genetic counseling. J Clin
Casanueva F, Finn JD, Forti G, Pinsky L (1995) Evidence for a Endocrinol Metab 90:106111
Giwercman A, Han TS, Kula K, repressive function of the long
Lean ME, Pendleton N, Punab M, Kooy RF, Reyniers E, Storm K, Vits L,
polyglutamine tract in the human van Velzen D, de Ruiter PE,
Silman AJ, Wu FC; European Male androgen receptor: possible
Ageing Study Group (2009) Brinkmann AO, de Paepe A,
pathogenetic relevance for the Willems PJ (1999) CAG repeat
Increased estrogen rather than (CAG)n-expanded neuronopathies.
decreased androgen action is contraction in the androgen
Hum Mol Genet 4:523527 receptor gene in three brothers with
associated with longer androgen
receptor CAG repeats. J Clin Kennedy WR, Alter M, Sung JH (1968) mental retardation. Am J Med Gen
Endocrinol Metab 94:277284 Progressive proximal spinal and 85:209213

53
Chapter 3: Pathophysiology of the androgen receptor

Kubini K, Zachmann M, Albers N, Lavery DN, McEwan IJ (2008a) Lundberg-Giwercman Y, Xu C,


Hiort O, Bettendorf M, Wolfle J, Functional characterization of the Arver S, Pousette A, Reneland R
Bidlingmaier F, Klingmuller D native NH2-terminal (1998) No association between the
(2000) Basal inhibin B and the transactivation domain of the androgen receptor gene CAG repeat
testosterone response to human human androgen receptor: binding and impaired sperm production in
chorionic gonadotropin correlate in kinetics for interactions with TFIIF Swedish men. Clin Genet
prepubertal boys. J Clin Endocrinol and SRC-1a. Biochemistry 54:435436
Metab 85:134138 47:33523359 Mahendroo MS, Cala KM, Hess DL,
Kuhlenbumer G, Kress W, Ringelstein Lavery DN, McEwan IJ (2008b) Russell DW (2001) Unexpected
EB, Stgbauer F (2001) Thirty-seven Structural characterization of the virilization in male mice lacking
CAG repeats in the androgen native NH2-terminal steroid 5 alpha-reductase enzymes.
receptor gene in two healthy transactivation domain of the Endocrinology 142:46524662
individuals. J Neurol 248:2326 human androgen receptor: a
Malki S, Nef S, Notarnicola C,
Kurzrock EA, Baskin LS, Li Y, collapsed disordered conformation
Thevenet L, Gasca S, Mejean C,
Cunha GR (1999) Epithelial- underlies structural plasticity and
Berta P, Poulat F, Boizet-Bonhoure
mesenchymal interactions in protein-induced folding.
B (2005) Prostaglandin D2 induces
development of the mouse fetal Biochemistry 47:33603369
nuclear import of the sex-
genital tubercle. Cells Tissues Organs Legius E, Vanderschueren D, Spiessens determining factor SOX9 via its
164:125130 C, DHooghe T, Matthijs G (1999) cAMP-PKA phosphorylation.
LaMarca HL, Rosen JM (2008) Association between CAG repeat EMBO J 24:17981809
Minireview: hormones and number in the androgen receptor
Mangelsdorf DJ, Thummel C, Beato M,
mammary cell fate what will and male infertility in a Belgian
Herrlich P, Schutz G, Umesono K,
I become when I grow up? study. Clin Genet 56:166167
Blumberg B, Kastner P, Mark M,
Endocrinology 149:43174321 Limer KL, Pye SR, Thomson W, Chambon P, Evans RM (1995) The
Lasnitzki I, Mizuno T (1977) Induction Boonen S, Borghs H, nuclear receptor superfamily: the
of the rat prostate gland by Vanderschueren D, Huhtaniemi IT, second decade. Cell 83:835839
androgens in organ culture. Adams JE, Ward KA, Platt H, Payne
Mariotti C, Castellotti B, Pareyson D,
J Endocrinol 74:4755 D, John SL, Bartfai G, Casanueva F,
Testa D, Eoli M, Antozzi C, Silani
Finn JD, Forti G, Giwercman A,
Lasnitzki I, Mizuno T (1979) Role of V, Marconi R, Tezzon F, Siciliano
Han TS, Kula K, Lean ME,
the mesenchyme in the induction of G, Marchini C, Gellera C, Donato
Pendleton N, Punab M, Silman AJ,
the rat prostate gland by androgens SD (2000) Phenotypic
Wu FC, ONeill TW; EMAS Study
in organ culture. J Endocrinol manifestations associated with
Group (2009) Genetic variation in
82:171178 sex hormone genes influences heel
CAG-repeat expansion in the
androgen receptor gene in male
La Spada AR, Wilson EM, Lubahn DB, ultrasound parameters in middle-
patients and heterozygous females: a
Harding AE, Fischbeck KH (1991) aged and elderly men: results from
clinical and molecular study of 30
Androgen receptor gene mutations the European Male Aging Study
families. Neuromusc Dis 10:391397
in X-linked spinal and bulbar (EMAS). J Bone Miner Res
muscular atrophy. Nature 24:314323 Marker PC, Donjacour AA, Dahiya R,
352:7779 Lubahn DB, Joseph DR, Sullivan PM, Cunha GR (2003) Hormonal,
Willard HF, French FS, Wilson EM cellular, and molecular control of
Latil AG, Azzouzi R, Cancel GS,
(1988) Cloning of human androgen prostatic development. Dev Biol
Guillaume EC, Cochan-Priollet B,
receptor complementary DNA and 253:165174
Berthon PL, Cussenot O (2001)
Prostate carcinoma risk and allelic localization to the X chromosome. Marshall GR, Wickings EJ, Nieschlag E
variants of genes involved in Science 240:327330 (1984) Testosterone can initiate
androgen biosynthesis and Lubahn DB, Brown TR, Simental JA, spermatogenesis in an immature
metabolism pathways. Cancer Higgs HN, Migeon CJ, Wilson EM, nonhuman primate, Macaca
92:11301137 French FS (1989) Sequence of the fascicularis. Endocrinology
Lavery DN, McEwan IJ (2006) The intron/exon junctions of the coding 114:22282233
human androgen receptor AF1 region of the human androgen Matias PM, Donner P, Coelho R,
transactivation domain: interactions receptor gene and identification of a Thomaz M, Peixoto C, Macedo S,
with transcription factor IIF and point mutation in a family with Otto N, Joschko S, Scholz P, Wegg
molten-globule-like structural complete androgen insensitivity. A, Basler S, Schafer M, Egner U,
characteristics. Biochem Soc Trans Proc Natl Acad Sci USA Carrondo MA (2000) Structural
34:10541057 86:95349538 evidence for ligand specificity in the

54
Chapter 3: Pathophysiology of the androgen receptor

binding domain of the human Moras D, Gronemeyer H (1998) The University of Washington, Seattle
androgen receptor. Implications for nuclear receptor ligand-binding [updated Sep 15, 2009]
pathogenic gene mutations. J Biol domain: structure and function. Page ST, Kupelian V, Bremner WJ,
Chem 275:2616426171 Curr Opin Cell Biol 10:384391 McKinlay JB (2006) The androgen
Meeks JJ, Schaeffer EM (2011) Genetic Murakami R, Mizuno T (1986) receptor gene CAG repeat
regulation of prostate development. Proximal-distal sequence of polymorphism does not predict
J Androl 32:210217 development of the skeletal tissues increased risk of heart disease:
Mengual L, Oriola J, Ascaso C, Ballesca in the penis of rat and the inductive longitudinal results from the
JL, Oliva R (2003) An increased effect of epithelium. J Embryol Exp Massachusetts Male Ageing Study.
CAG repeat length in the androgen Morphol 92:133143 Clin Endocrinol (Oxf) 65:333339
receptor gene in azoospermic ICSI Nielsen TL, Hagen C, Wraae K, Papadimitriou DT, Linglart A, Morel
candidates. J Androl 24:279284 Bathum L, Larsen R, Brixen K, Y, Chaussain JL (2006) Puberty in
Mifsud A, Sim CK, Boettger-Tong H, Andersen M (2010) The impact of subjects with complete androgen
Moreira S, Lamb DJ, Lipshultz LI, the CAG repeat polymorphism of insensitivity syndrome. Horm Res
Yong EL (2001) Trinucleotide the androgen receptor gene on 65:126131
(CAG) repeat polymorphisms in the muscle and adipose tissues in 20 Patrizio P, Leonard DG, Chen KL,
androgen receptor gene: molecular 29-year-old Danish men: Odense Hernandez-Ayup S, Trounson AO
markers of risk for male infertility. Androgen Study. Eur J Endocrinol (2001) Larger trinucleotide repeat
Fertil Steril 75:275281 162:795804 size in the androgen receptor gene
Mitsumori K, Terai A, Oka H, Segawa Nishida H, Miyagawa S, Matsumaru D, of infertile men with extremely
T, Ogura K, Yoshida O, Ogawa O Wada Y, Satoh Y, Ogino Y, Fukuda severe oligozoospermia. J Androl
(1999) Androgen receptor CAG S, Iguchi T, Yamada G (2008) Gene 22:444448
repeat length polymorphism in expression analyses on embryonic Perriton CL, Powles N, Chiang C,
benign prostatic hyperplasia (BPH): external genitalia: identification of Maconochie MK, Cohn MJ (2002)
correlation with adenoma growth. regulatory genes possibly involved Sonic hedgehog signaling from the
Prostate 41:253257 in masculinization processes. urethral epithelium controls
Miyagawa S, Satoh Y, Haraguchi R, Congenit Anom (Kyoto) 48:6367 external genital development. Dev
Suzuki K, Iguchi T, Taketo MM, OShaughnessy PJ, Baker PJ, Heikkila Biol 247:2646
Nakagata N, Matsumoto T, M, Vainio S, McMahon AP (2000) Pioro EP, Kant A, Mitsumoto H (1994)
Takeyama K, Kato S, Yamada G Localization of 17beta- Disease expression in a Kennedys
(2009) Genetic interactions of the hydroxysteroid dehydrogenase/17- disease kindred is unrelated to CAG
androgen and Wnt/beta-catenin ketosteroid reductase isoform tandem repeat size in the androgen
pathways for the masculinization of expression in the developing mouse receptor gene: characterization in a
external genitalia. Mol Endocrinol testis androstenedione is the symptomatic female. Ann Neurol
23:871880 major androgen secreted by fetal/ 36:318
Miyagawa S, Matsumaru D, neonatal Leydig cells. Endocrinology Piprek RP (2009) Genetic mechanisms
Murashima A, Omori A, Satoh Y, 141:26312637 underlying male sex determination
Haraguchi R, Motoyama J, Iguchi T, OShaughnessy PJ, Johnston H, in mammals. J Appl Genet
Nakagata N, Hui CC, Yamada G Willerton L, Baker PJ (2002) Failure 50:347360
(2011) The role of sonic hedgehog- of normal adult Leydig cell Plant TM (2006) The role of KiSS-1 in
gli2 pathway in the masculinization development in androgen-receptor- the regulation of puberty in higher
of external genitalia. Endocrinology deficient mice. J Cell Sci primates. Eur J Endocrinol 155
152:28942903 115:34913496 (Suppl 1):S11S16
Moniot B, Declosmenil F, Barrionuevo OShaughnessy PJ, Baker PJ, Johnston Plant TM, Witchel SF (2006) Puberty
F, Scherer G, Aritake K, Malki S, H (2006) The foetal Leydig cell in non-human primates and
Marzi L, Cohen-Solal A, Georg I, differentiation, function and humans. In: Neill JD, Plant TM,
Klattig J, Englert C, Kim Y, Capel B, regulation. Int J Androl 29:9095; Pfaff DW, Challis JRG, de Kretser
Eguchi N, Urade Y, Boizet- discussion:105108 DM, Richards JS, Wassarman PM
Bonhoure B, Poulat F (2009) The
Ostrer H (2008) 46,XY disorder of sex (eds) Knobil and Neills Physiology of
PGD2 pathway, independently of
development and 46,XY complete Reproduction. Academic Press,
FGF9, amplifies SOX9 activity in
gonadal dysgenesis. In: Pagon RA, St Louis, pp 21772230
Sertoli cells during male sexual
differentiation. Development Bird TD, Dolan CR, Stephens K Platz EA, Giovannucci E, Dahl DM,
136:18131821 (eds) GeneReviews [Internet]. Krithivas K, Hennekens CH, Brown

55
Chapter 3: Pathophysiology of the androgen receptor

M, Stampfer MJ, Kantoff PW (1998) Rey RA, Belville C, Nihoul-Fekete C, estrogen receptor gene
The androgen receptor gene GGN Michel-Calemard L, Forest MG, polymorphism in men. Anthropol
microsatellite and prostate cancer Lahlou N, Jaubert F, Mowszowicz I, Anzeiger 59:343353
risk. Cancer Epidemiol Biomarkers David M, Saka N, Bouvattier C, Sasagawa I, Suzuki Y, Ashida J, Nakada
Prev 7:379384. Bertrand AM, Lecointre C, Soskin S, T, Muroya K, Ogata T (2001) CAG
Platz EA, Rimm EB, Willett WC, Cabrol S, Crosnier H, Leger J, repeat length analysis and mutation
Kantoff PW, Giovannucci E (2000) Lortat-Jacob S, Nicolino M, Rabl W, screening of the androgen receptor
Racial variation in prostate cancer Toledo SP, Bas F, Gompel A, gene in Japanese men with
incidence and in hormonal system Czernichow P, Chatelain P, idiopathic azoospermia. J Androl
markers among male health Rappaport R, Morel Y, Josso N 22:804808
professionals. J Natl Cancer Inst (1999) Evaluation of gonadal
function in 107 intersex patients by Sawaya ME, Shalita AR (1998)
92:20092017 Androgen receptor polymorphisms
means of serum antimullerian
Podlasek CA, Barnett DH, Clemens JQ, hormone measurement. J Clin (CAG repeat lengths) in
Bak PM, Bushman W (1999) Endocrinol Metab 84:627631 androgenetic alopecia hirsutism and
Prostate development requires acne. J Cutan Med Surg 3:915
Rodien P, Mebarki F, Mowszowicz I,
Sonic hedgehog expressed by the Schauwaers K, De Gendt K, Saunders
Chaussain JL, Young J, Morel Y,
urogenital sinus epithelium. Dev PT, Atanassova N, Haelens A,
Schaison G (1996) Different
Biol 209:2839 Callewaert L, Moehren U, Swinnen
phenotypes in a family with
Pope HG, Cohane GH, Kanayama G, androgen insensitivity caused by the JV, Verhoeven G, Verrijdt G,
Siegel AJ, Hudson JI (2003) same M780I point mutation in the Claessens F (2007) Loss of androgen
Testosterone gel supplementation androgen receptor gene. J Clin receptor binding to selective
for men with refrectory Endocrinol Metab 81:29942998 androgen response elements causes
depression: a randomized placebo- a reproductive phenotype in a
Ross RK, Pike MC, Coetzee GA, knockin mouse model. Proc Natl
controlled trial. Am J Psychiatry
Reichardt JK, Yu MC, Feigelson H, Acad Sci USA 104:49614966
160:105111
Stanczyk FZ, Kolonel LN,
Price H, McNeal JE, Stamey TA (1990) Henderson BE (1998) Androgen Schneider G, Nienhaus K, Gromoll J,
Evolving patterns of tissue metabolism and Prostate Cancer: Heuft G, Nieschlag E, Zitzmann M
composition in benign prostatic establishing a model of genetic (2011a) Depressive symptoms in
hyperplasia as a function of susceptibility. Cancer Res men aged 50 years and older and
specimen size. Hum Pathol 58:44974504 their relationship to genetic
21:578585 androgen receptor polymorphism
Sack JS, Kish KF, Wang C, Attar RM, and sex hormone levels in three
Quigley CA, De Bellis A, Marschke KB, Kiefer SE, An Y, Wu GY, Scheffler different samples. Am J Geriatr
el-Awady MK, Wilson EM, French JE, Salvati ME, Krystek SR Jr, Psychiatry 19:274283
FS (1995) Androgen receptor Weinmann R, Einspahr HM (2001)
defects: historical, clinical, and Crystallographic structures of the Schneider G, Nienhaus K, Gromoll J,
molecular perspectives. Endocr Rev ligand-binding domains of the Heuft G, Nieschlag E, Zitzmann M
16:271321 androgen receptor and its T877A (2011b) Sex hormone levels, genetic
mutant complexed with the natural androgen receptor polymorphism,
Rajpert-De Meyts E, Leffers H, and anxiety in 50-year-old males.
Petersen JH, Andersen AG, Carlsen agonist dihydrotestosterone. Proc
Natl Acad Sci USA 98:49044909 J Sex Med 8:34523464
E, Jorgensen N, Skakkebaek NE
(2002) CAG repeat length in Salmen T, Heikkinen AM, Mahonen A, Schweiger U, Deuschle M, Weber B,
androgen-receptor gene and Kroger H, Komulainen M, Pallonen Korner A, Lammers CH, Schmider J
reproductive variables in fertile and H, Saarikoski S, Honkanen R, (1999) Testosterone gonadotropin
infertile men. Lancet 359:4446 Maenpaa PH (2003) Relation of and cortisol secretion in male
androgen receptor gene patients with major depression.
Remes T, Vaisanen SB, Mahonen A, Psychosom Med 61:292296
Huuskonen J, Kroger H, Jurvelin JS, polymorphism to bone mineral
Penttila IM, Rauramaa R (2003) density and fracture risk in early Seidman SN, Araujo AB, Roose SP,
Aerobic exercise and bone mineral postmenopausal women during a 5- McKinlay JB (2001) Testosterone
density in middle-aged Finnish year randomized hormone level androgen receptor
men: a controlled randomized trial replacement therapy trial. J Bone polymorphism and depressive
with reference to androgen receptor Miner Res 18:319324 symptoms in middle-aged men. Biol
aromatase and estrogen receptor Sapir-Koren R, Livshits G, Landsman Psych 50:371376
alpha gene polymorphisms. Bone T, Kobyliansky E (2001) Bone Seifert AW, Zheng Z, Ormerod BK,
32:412420 mineral density is associated with Cohn MJ (2010) Sonic hedgehog

56
Chapter 3: Pathophysiology of the androgen receptor

controls growth of external genitalia M, Jannausch M (1999) Genetic Biochem Biophys Res Commun
by regulating cell cycle kinetics. Nat markers, bone mineral density, and 153:241248
Commun 1:23 serum osteocalcin levels. J Bone Min Tut TG, Ghadessy FJ, Trifiro MA,
Res 14:14111419 Pinsky L, Yong EL (1997) Long
Sekido R, Lovell-Badge R (2008) Sex
determination involves synergistic Stanford JL, Just JJ, Gibbs M, Wicklund polyglutamine tracts in the
action of SRY and SF1 on a specific KG, Neal CL, Blumenstein BA, androgen receptor are associated
Sox9 enhancer. Nature 453:930934 Ostrander EA (1997) Polymorphic with reduced trans-activation
repeats in the androgen receptor impaired sperm production and
Sekido R, Lovell-Badge R (2009) Sex
gene: molecular markers of prostate male infertility. J Clin Endocr Metab
determination and SRY: down to a
cancer risk. Cancer Res 82:37773782
wink and a nudge? Trends Genet
57:11941198
25:1929 Unden F, Ljunggren JG, Beck-Friis J,
Stanworth RD, Kapoor D, Channer KS, Kjellman F, Wetterberg L (1988)
Shabsigh R, Arver S, Channer KS, Jones TH (2008) Androgen receptor Hypothalamic-pituitary-gonadal
Eardley I, Fabbri A, Gooren L, CAG repeat polymorphism is axis in major depressive disorders.
Heufelder A, Jones H, Meryn S, associated with serum testosterone Acta Psychiatri Scand 78:138146
Zitzmann M (2008) The triad of levels, obesity and serum leptin in
erectile dysfunction, hypogonadism van Golde R, Van Houwelingen K,
men with type 2 diabetes. Eur Kiemeney L, Kremer J, Tuerlings J,
and the metabolic syndrome. Int J Endocrinol 159:739746
J Clin Pract 62:791798 Schalken J, Meuleman E (2002) Is
Steltenkamp S, Hiort O (2007) Pubertal increased CAG repeat length in the
Shaffer PL, Jivan A, Dollins DE, development of 46,XY DSD patients androgen receptor gene a risk factor
Claessens F, Gewirth DT (2004) with partial androgen insensitivity for male subfertility? J Urol
Structural basis of androgen due to mutation of the androgen 167:621623
receptor binding to selective receptor assigned to male sex. Horm
androgen response elements. Proc van Houten ME, Gooren LJ (2000)
Res 68:205 Differences in reproductive
Natl Acad Sci USA 101:47584763
Taguchi O, Cunha GR, Lawrence WD, endocrinology between Asian men
Shen JH, Ingraham HA (2002) Robboy SJ (1984) Timing and and Caucasian mena literature
Regulation of the orphan nuclear irreversibility of Mullerian duct review. Asian J Androl 2:1320
receptor steroidogenic factor 1 by inhibition in the embryonic Van Pottelbergh I, Lumbroso S,
Sox proteins. Mol Endocrinol reproductive tract of the human Goemaere S, Sultan C, Kaufman JM
16:529540 male. Dev Biol 106:394398 (2001) Lack of influence of the
Simental JA, Sar M, Lane MV, French Takeda H, Lasnitzki I, Mizuno T (1986) androgen receptor gene CAG-repeat
FS, Wilson EM (1991) Analysis of prostatic bud induction polymorphism on sex steroid status
Transcriptional activation and by brief androgen treatment in the and bone metabolism in elderly
nuclear targeting signals of the fetal rat urogenital sinus. men. Clin Endocrinol 55:659666
human androgen receptor. J Biol J Endocrinol 110:467470 von Eckardstein S, Syska A, Gromoll J,
Chem 266:510518
Thomas MA, Preece DM, Bentel JM Kamischke A, Simoni M, Nieschlag
Sinnecker GH, Hiort O, Nitsche EM, (2010) Androgen regulation of the E (2001) Inverse correlation
Holterhus PM, Kruse K (1997) prostatic tumour suppressor between sperm concentration and
Functional assessment and clinical NKX3.1 is mediated by its 30 number of androgen receptor CAG
classification of androgen sensitivity untranslated region. Biochem J repeats in normal men. J Clin
in patients with mutations of the 425:575583 Endocr Metab 86:25852590
androgen receptor gene. German
Tilley WD, Marcelli M, Wilson JD, von Eckardstein S, Schmidt A,
Collaborative Intersex Study Group.
McPhaul MJ (1989) Kamischke A, Simoni M, Gromoll J,
Eur J Pediatr 156:714
Characterization and expression of Nieschlag E (2002) CAG repeat
Song LN, Herrell R, Byers S, Shah S, a cDNA encoding the human length in the androgen receptor
Wilson EM, Gelmann EP (2003) androgen receptor. Proc Natl Acad gene and gonadotrophin
Beta-catenin binds to the activation Sci USA 86:327331 suppression influence the
function 2 region of the androgen Trapman J, Klaassen P, Kuiper GG, van effectiveness of hormonal male
receptor and modulates the effects der Korput JA, Faber PW, van Rooij contraception. Clin Endocrinol
of the N-terminal domain and TIF2 HC, Geurts van Kessel A, Voorhorst 57:647655
on ligand-dependent transcription. MM, Mulder E, Brinkmann AO Wallerand H, Remy-Martin A,
Mol Cell Biol 23:16741687 (1988) Cloning, structure and Chabannes E, Bermont L, Adessi
Sowers M, Willing M, Burns T, expression of a cDNA encoding GL, Bittard H (2001) Relationship
Deschenes S, Hollis B, Crutchfield the human androgen receptor. between expansion of the CAG

57
Chapter 3: Pathophysiology of the androgen receptor

repeat in exon 1 of the androgen glycine repeats. J Clin Endocrinol signaling specifies fetal Leydig cell
receptor gene and idiopathic male Metab 91:35153520 fate in testis organogenesis. Genes
infertility. Fertil Steril 76:769774 Werner R, Schutt J, Hannema S, Ropke Dev 16:14331440
Wang C, Nieschlag E, Swerdloff R, A, Wieacker P, Hiort O, Holterhus Yeh S, Tsai MY, Xu Q, Mu XM, Lardy
Behre HM, Hellstrom WJ, Gooren PM (2006b) Androgen receptor H, Huang KE, Lin H, Yeh SD,
LJ, Kaufman JM, Legros JJ, gene mutations in androgen Altuwaijri S, Zhou X, Xing L, Boyce
Lunenfeld B, Morales A, Morley JE, insensitivity syndrome cause BF, Hung MC, Zhang S, Gan L,
Schulman C, Thompson IM, distinct patterns of reduced Chang C (2002) Generation and
Weidner W, Wu FC (2008) activation of androgen-responsive characterization of androgen
Investigation, treatment and promoter constructs. J Steroid receptor knockout (ARKO) mice:
monitoring of late-onset Biochem Mol Biol 101:110 an in vivo model for the study of
hypogonadism in males: ISA, Werner R, Grotsch H, Hiort O (2010) 46, androgen functions in selective
ISSAM, EAU, EAA and ASA XY disorders of sex development tissues. Proc Natl Acad Sci USA
recommendations. Eur J Endocrinol the undermasculinised male with 99:1349813503
159:507514 disorders of androgen action. Best Yong EL, Lim LS, Wang Q, Mifsud A,
Wang G, Chen G, Wang X, Zhong J, Lu Pract Res Clin Endocrinol Metab Lim J, Ong YC, Sim KS (2000)
J (2001) The polymorphism of 24:263277 Androgen receptor polymorphisms
(CAG)n repeats within androgen Wilhelm D, Martinson F, Bradford S, and mutations in male infertility.
receptor gene among Chinese male Wilson MJ, Combes AN, Beverdam J Endocrinol Invest 23:573577
population [article in Chinese]. A, Bowles J, Mizusaki H, Koopman Yoshida KI, Yano M, Chiba K, Honda
Zhonghua Yi Xue Yi Chuan Xue Za P (2005) Sertoli cell differentiation M, Kitahara S (1999) CAG repeat
Zhi [Chinese Journal of Medical is induced both cell-autonomously length in the androgen receptor
Genetics] 18:456458 and through prostaglandin gene is enhanced in patients with
Wang Q, Udayakumar TS, Vasaitis TS, signaling during mammalian sex idiopathic azoospermia. Urology
Brodie AM, Fondell JD (2004) determination. Dev Biol 54:10781081
Mechanistic relationship between 287:111124
Zeegers MP, Kiemeney LA, Nieder
androgen receptor polyglutamine Wilhelm D, Hiramatsu R, Mizusaki H, AM, Ostrer H (2004) How strong is
tract truncation and androgen- Widjaja L, Combes AN, Kanai Y, the association between CAG and
dependent transcriptional Koopman P (2007) SOX9 regulates GGN repeat length polymorphisms
hyperactivity in prostate prostaglandin D synthase gene in the androgen receptor gene and
cancer cells. J Biol Chem transcription in vivo to ensure testis prostate cancer risk? Cancer
279:1731917328 development. J Biol Chem Epidemiol Biomarkers Prev
Warot X, Fromental-Ramain C, 282:1055310560 13:17651771
Fraulob V, Chambon P, Dolle P Yamada G, Satoh Y, Baskin LS, Cunha Zeltser L, Desplan C, Heintz N (1996)
(1997) Gene dosage-dependent GR (2003) Cellular and molecular Hoxb-13: a new Hox gene in a
effects of the Hoxa-13 and Hoxd-13 mechanisms of development of the distant region of the HOXB cluster
mutations on morphogenesis of the external genitalia. Differentiation maintains colinearity. Development
terminal parts of the digestive and 71:445460 122:24752484
urogenital tracts. Development Yamada G, Suzuki K, Haraguchi R,
124:47814791 Zhou X (2010) Roles of androgen
Miyagawa S, Satoh Y, Kamimura M, receptor in male and female
Welsh M, MacLeod DJ, Walker M, Nakagata N, Kataoka H, Kuroiwa A, reproduction: lessons from global
Smith LB, Sharpe RM (2009) Chen Y (2006) Molecular genetic and cell-specific androgen receptor
Critical androgen-sensitive periods cascades for external genitalia knockout (ARKO) mice. J Androl
of rat penis and clitoris formation: an emerging 31:235243
development. Int J Androl 33: organogenesis program. Dev Dyn
235:17381752 Zhou ZX, Sar M, Simental JA, Lane
e144e152
MV, Wilson EM (1994) A ligand-
Werner R, Holterhus PM, Binder G, Yanovich R, Milgrom R, Friedman E, dependent bipartite nuclear
Schwarz HP, Morlot M, Struve D, Moran DS (2011) Androgen targeting signal in the human
Marschke C, Hiort O (2006a) The receptor CAG repeat size is androgen receptor. Requirement
A645D mutation in the hinge associated with stress fracture risk: a for the DNA-binding domain
region of the human androgen pilot study. Clin Orthop Relat Res and modulation by NH2-terminal
receptor (AR) gene modulates AR 469:29252931 and carboxyl-terminal
activity, depending on the context of Yao HH, Whoriskey W, Capel B (2002) sequences. J Biol Chem
the polymorphic glutamine and Desert Hedgehog/Patched 1 269:1311513123

58
Chapter 3: Pathophysiology of the androgen receptor

Zitzmann M, Nieschlag E (2001) Zitzmann M, Brune M, Nieschlag E substituted hypogonadal men are
Testosterone levels in healthy men (2002a) Vascular reactivity in dependent on the CAG repeat
and the relation to behavioural and hypogonadal men is reduced by polymorphism of the androgen
physical characteristics: facts and androgen substitution. J Clin Endocr receptor gene: a longitudinal
constructs. Euro J Endocrinol Metab 87:50305037 pharmacogenetic study. J Clin
144:183197 Endocrinol Metab 88:20492054
Zitzmann M, Brune M, Vieth V,
Zitzmann M, Brune M, Kornmann B, Nieschlag E (2002b) Monitoring Zmuda JM, Cauley JA, Kuller LH,
Gromoll J, Junker R, Nieschlag E bone density in hypogonadal men Newman AB, Robbins J, Harris T,
(2001a) The CAG repeat by quantitative phalangeal Ferrell RE (2000a) Androgen
polymorphism in the androgen ultrasound. Bone 31:422429 receptor CAG repeat
receptor gene affects bone density polymorphism: a novel marker of
and bone metabolism in healthy Zitzmann M, Gromoll J, von osteoporotic risk in men.
males. Clin Endocrinol 55:649657 Eckardstein A, Nieschlag E (2003a) Osteoporosis Int 11 S151: 355
The CAG repeat polymorphism in
Zitzmann M, Brune M, Kornmann B, the androgen receptor gene Zmuda JM, Cauley JA, Kuller LH,
Gromoll J, von Eckardstein S, von modulates body fat mass and serum Zhang X, Palermo L, Nevitt MC,
Eckardstein A, Nieschlag E (2001b) levels of leptin and insulin in men. Ferrell RE (2000b) Androgen
The CAG repeat polymorphism in Diabetologia 46:3139 receptor CAG repeat length is
the AR gene affects high density associated with increased hip bone
lipoprotein cholesterol and arterial Zitzmann M, Depenbusch M, Gromoll loss and vertebral fracture risk
vasoreactivity. J Clin Endocr Metab J, Nieschlag E (2003b) Prostate among older men. J Bone Min Res
86:48674873 volume and growth in testosterone- 15 S491: M141

59
Chapter
Methodology for measuring testosterone,

4 dihydrotestosterone and sex hormone-


binding globulin in a clinical setting
Manuela Simoni, Flaminia Fanelli, Laura Roli, and Uberto Pagotto

4.1 Introduction 60 4.3.4.2 Ultrafiltration 76


4.2 Testosterone, dihydrotestosterone and sex 4.3.4.3 Direct free testosterone assays 76
hormone-binding globulin in blood 60 4.3.4.4 Salivary testosterone 77
4.3 Measurement of testosterone 62 4.3.4.5 Bioavailable testosterone 77
4.3.1 Mass spectrometry methods 63 4.3.4.6 Calculated free testosterone 78
4.3.1.1 Gas chromatographymass 4.3.5 Bioassay 78
spectrometry 63 4.4 Measurement of dihydrotestosterone 78
4.3.1.2 Liquid chromatographytandem 4.5 Quality control 79
mass spectrometry 64 4.5.1 Choice of the kit and assay
4.3.1.3 Comparative analytical performance validation 79
and standardization 65 4.5.2 Internal quality control 80
4.3.2 Radioimmunoassay 73 4.5.3 External quality assessment 81
4.3.3 Other immunoassays 74 4.6 Key messages 82
4.3.4 Assessment of free testosterone 75 4.7 References 82
4.3.4.1 Equilibrium dialysis 75

4.1 Introduction 4.2 Testosterone,


Testosterone is a hormone difficult to measure accur-
ately. Yet, its accurate determination is the prerequis-
dihydrotestosterone and sex
ite for the correct diagnosis and clinical management hormone-binding globulin in blood
of hypogonadism in males and hyperandrogenism in Testosterone and DHT circulate in serum largely
females. In the last decade a number of studies bound to transport proteins: that is albumin, which
increased awareness of the poor performance of most displays low affinity but very high binding capacity,
of the current assays and identified some strategies to and SHBG, with high affinity and low capacity.
improve the accuracy of testosterone testing (Rosner A systematic analysis of serum transport of steroid
et al. 2007; 2010). In the previous edition of this book hormones and their interaction with binding proteins
a chapter was dedicated to the description of the revealed an association constant of SHBG of 1.6  109
principles, analytical performance and limitations of M1 for testosterone and of 5.5  109 M1 for DHT
the existing methodologies for measuring testoster- at 37  C (Dunn et al. 1981). By comparison the
one, DHT and SHBG (Simoni 2004). Here, we will association constant of albumin for testosterone is
provide an update on the state of the art to help the five orders of magnitude lower (6  104 M1)
reader choose the testosterone detection system most (Anderson 1974). The relative amounts of protein
suitable for his/her needs in view of the current binding of circulating testosterone in men and
recommendations. women are shown in Table 4.1.

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

60
Chapter 4: Measuring testosterone, DHT and SHBG

Table 4.1 Transport of endogenous testosterone and DHT in male and female serum

Serum concentration (nM) Unbound (%) SHBG (%) CBG (%) Albumin (%)
Testosterone
Adult men 23.0 2.23 44.3 3.56 49.9
Adult women
Follicular phase 1.3 1.36 66.0 2.26 30.4
Luteal phase 1.3 1.37 65.7 2.20 30.7
Pregnancy 4.7 0.23 95.4 0.82 3.6
DHT
Adult men 1.70 0.88 49.7 0.22 39.2
Adult women
Follicular phase 0.65 0.47 78.4 0.12 21.0
Luteal phase 0.65 0.48 78.1 0.12 21.3
Pregnancy 0.93 0.07 97.8 0.04 21.2
Source: Dunn et al. (1981).
Abbreviations: CBG, cortisol-binding globulin; SHBG, sex hormone-binding globulin; DHT, dihydrotestosterone.

About 1.52% of serum testosterone is free and is The free-hormone hypothesis has been repeat-
believed to represent bioactive testosterone. Free and edly challenged in the scientific literature, mainly due
protein-bound testosterone and DHT are in equilib- to the difficulty of reconciling the existing experimen-
rium, so that when free hormone is subtracted from tal evidence with appropriate mathematical models of
circulation because of entry into tissue, new testoster- hormone transport (Ekins 1990; Mendel 1992). For
one dissociates from albumin and SHBG, a new equi- instance, the low affinity of testosterone for albumin
librium is promptly reached, and the free-hormone binding and some experimental data led to the idea
concentration in serum remains constant. Conversely, that albumin-bound testosterone is readily available
pathophysiological conditions causing changes in for delivery to the tissues (i.e. bioavailable); while only
binding protein concentration (e.g. pregnancy, hypo- SHBG-bound testosterone is not biologically active
or hyperthyroidism, growth hormone (GH) excess, (Manni et al. 1985). In contrast, SHBG itself has been
treatment with antiepileptic drugs) or displacement proposed to interact with cell surface receptors,
of testosterone from SHBG by drugs (e.g. danazol) thereby contributing to the biological activity of
results in changes in total testosterone concentration androgens (Rosner et al. 1999). This novel, putative
in order to maintain constant free testosterone levels. function of SHBG is of particular interest in the light
The measurement of SHBG is valuable for assess- of the essential lack of any physiological explanation
ment of androgenization and of free testosterone. In of why primates, unlike all other species, possess such
earlier times SHBG was measured indirectly, by esti- a protein. Sex hormone-binding globulin seems to
mating its binding capacity. The classic method used buffer serum testosterone levels, which, beside the
tritiated DHT as ligand because of its higher affinity physiological circadian rhythm, show only minor cir-
to SHBG and lack of binding to cortisol-binding choral variations despite highly pulsatile LH secretion
globulin (CBG). Saturating amounts of labeled DHT (Simoni et al. 1988; 1992). In contrast, serum testos-
were added to the samples and SHBG was then pre- terone levels oscillate widely in rodents, which do not
cipitated by ammonium sulfate. The amount of have SHBG. In addition SHBG reduces the rate of
labeled DHT precipitated provided a direct measure- hepatic testosterone degradation. There are no known
ment of SHBG binding capacity. This method did not cases of congenital absence of SHBG in humans, but
allow absolute changes in SHBG protein concentra- an analbuminemic strain of rats, a species which
tions to be measured, which can now be assessed by does not have circulating SHBG, is normally fertile
modern immunoradiometric assays. Modern assays and shows normal free testosterone levels, arguing
have demonstrated that, in general, SHBG binding for a dispensable role of serum-testosterone-binding
capacity (expressed in terms of DHT binding) corres- proteins (Mendel et al. 1989). Similarly, the congeni-
ponds acceptably to the molar SHBG concentration. tal absence of thyroxin-binding globulin (TBG) in

61
Chapter 4: Measuring testosterone, DHT and SHBG

humans is compatible with normal thyroid function showing the greatest inaccuracy, as demonstrated by
(Dussault et al. 1977). proficiency tests results (Wang et al. 2004; Rosner
Several factors influence SHBG production and, et al. 2007). Testosterone detection in pediatric and
thereby, free testosterone levels. Estrogens stimulate female samples is affected by the physiologically low
and androgens inhibit SHBG secretion. Administra- concentrations of the hormone, the high concentra-
tion of 20 mg daily of ethinyl estradiol to men for five tions of other, interfering, cross-reacting steroids and
weeks resulted in a 150% increase in SHBG and, as a the age-related variability of matrix and steroid com-
consequence of the reduced free testosterone levels, in position (Rauh 2010). As a consequence, no trust-
a 50% increase in total serum testosterone (Anderson worthy age- and sex-specific reference ranges are
1974). The estrogen effect is responsible for the higher presently available for this important biomarker. Even
SHBG serum levels in women compared to men. In in adult males the current reference ranges contain a
pregnancy, SHBG rises to levels 510 times higher remarkable gray zone at about 812 nmol/l, leaving
than in non-pregnant women. In addition, SHBG the lowest level of normality basically undefined.
levels are stimulated by thyroid hormones, resulting Facing these problems, The Endocrine Society,
in high levels in thyrotoxycosis and low levels in in partnership with the Centers for Disease Control
hypothyroidism, and are reduced by GH and cortisol, and Prevention and other endorsing organizations,
resulting in low levels in acromegaly and in Cushing published a consensus statement in October 2010 with
syndrome. Finally, SHBG levels are higher in children the aim of stimulating the cooperation of clinical
than in adults and increase in men after the age of 50, laboratories, instrument makers, assay manufacturers,
contributing to the possible decline of free testoster- medical communities and other stakeholders in thor-
one levels observed in aging men. oughly reviewing the process, from guidelines for
The most important bioactive metabolite of tes- patient preparation and specimen handling to a work-
tosterone is DHT. The reduction of testosterone to able standard for the testosterone assay, the true core
DHT occurs in those tissues expressing 5a-reductase, target. This consensus statement contains seven recom-
and DHT is well measurable in circulation. In eugo- mendations to follow to achieve the goal of highly
nadal, adult men, serum DHT concentrations are accurate testosterone testing, and a timeline suggesting
about 1012 times lower than testosterone, and that standardization of testosterone assay could be
DHT is mainly bound to SHBG (Table 4.1). Given achieved by 2012 (Rosner et al. 2010). In summary
the role of DHT in prostate growth, the measurement these recommendations include: (1) all stakeholders
of serum DHT is of relevance during testosterone should work at testosterone assay standardization and
treatment, especially when testosterone is adminis- production of accurate and reliable tests; (2) experts
tered via the transdermal route (e.g. testosterone gel should work at defining performance criteria ensuring
or patches), since the skin is the primary organ for coverage of the full range of expected values; (3) refer-
5a-reduction. ence intervals in adults and children of both sexes
should be established; (4) guidelines for preanalytical
steps (patient preparation, specimen collection, etc.)
4.3 Measurement of testosterone should be prepared; (5) third-party payers and health-
Testosterone measurement is useful in the assessment care organizations should promote and reimburse
of a number of clinical conditions such as hypogo- only standardized, accuracy-based tests; (6) research
nadism, hyperandrogenism, diabetes, cardiovascular funding agencies and journals should support and
disease, bone diseases, neurovegetative disorders, publish, respectively, only research work based on
aging etc. (see Simoni and Nieschlag 2010), but, des- standardized testosterone tests; (7) manufacturers and
pite the importance of testosterone determination for laboratories should further develop new methods for
clinical decisions, the different methods in current use accurate, specific, sensitive and cost-effective testoster-
are frequently inaccurate and their results inconsist- one testing. Meanwhile everyone concerned should be
ent, especially at low concentrations. This problem is aware of the methods available and make an educated
mainly due to the lack of recognized reference stand- choice of the system that better suits his/her needs.
ards and is common to all types of assays currently A detailed list of the pros and cons of the current
available, from immunoassays to mass spectrometry methods has been summarized in this consensus state-
(MS) methods, with the automated immunoassays ment (Rosner et al. 2010).

62
Chapter 4: Measuring testosterone, DHT and SHBG

4.3.1 Mass spectrometry methods 4.3.1.1 Gas chromatographymass spectrometry


While testosterone immunoassays still dominate in In gas chromatography systems the analytes are
routine laboratories due to their indispensable high- injected into a capillary column filled with a silica
throughput capabilities, their shortcomings, espe- polymer. The analytes interact with the stationary
cially inaccuracy and poor reproducibility at low phase until the temperature, regulated by a heating
levels, prompted the advancement of a new gener- program, reaches the specific boiling point of the ana-
ation of MS technologies. lytes that hence become volatile and are carried to the
The application of gas chromatography combined detector by the gas flow. Since a very small number of
with mass spectrometry (GC-MS) to the discovery and molecules exhibit the thermo-stability and volatility
structural elucidation of steroid hormones and metab- features required by gas chromatography, strategies of
olites goes back to the 1960s, and this technique still chemical modification of the analytes prior to injection
represents not only the pre-eminent tool for steroid were developed: the so called derivatization. The most
disorder and metabolomic studies (Krone et al. 2010), common derivatives of testosterone are pentafluoro-
but also the gold standard and reference method used benzyloxime-trimethylsilyl-, t-butyldimethylsylil- and
for the validation of other assays. However, this tech- pentafluoropropionic anhydride (Fitzgerald et al.
nique did not overcome the need for an extensive 2010). The chemical modification is performed on the
sample pretreatment, thus preventing its introduction purified extract: after a liquid-liquid extraction (LLE)
to routine settings, and GC-MS survives as a preroga- involving solvents such as ethylacetate (Fitzgerald et al.
tive of only a few specialized laboratories. In contrast, 2010), diethylether (Taieb et al. 2003), dichloro-
the latest advances in the development of quantitative methane or hexane (Wolthers and Kraan 1999), a fur-
methods by liquid chromatographytandem mass ther clean-up is needed, and is usually accomplished by
spectrometry (LC-MS/MS), as witnessed by the expo- a gel chromatography on Sephadex LH-20 or water
nential number of publications on this topic since the extraction (Shackleton 2010). Before entering the mass
late 1990s, render LC-MS/MS potentially suitable for spectrometer, usually a single quadrupole, the analyte is
routine testosterone measurement, as it offers a prom- ionized by electronic impact, so that Gaussian-shaped
ising fusion of good accuracy and specificity, high- peaks of the whole molecule and of its specific frag-
throughput capabilities, and abatement of both direct ments, the fragmentography, appear at a specific reten-
and indirect costs of the present immunoassays. tion time, and are revealed for their mass-to-charge
In principle MS acts as a detector for the chroma- ratio (m/z). Usually one or two fragments, detected in
tographic technique to which it is combined, per- selected-ion monitoring mode, are used for the
forming, in addition, a further analysis based on the quantification.
recognition of intrinsic features of the molecules: The abundance of steps in the sample processing
molecular weight and structure. In quantitative appli- and analysis would inevitably result in high impreci-
cations, the measurement coincides with the identifi- sion of the assay. This is overcome by the use of
cation of the analyte, providing high specificity. stable, isotopically labeled internal standards, in
Chromatographic methods are based on the sep- which at least two atoms of hydrogen (H) or 12C are
aration of the molecules in a complex mixture replaced with deuterium (2H) or 13C, reducing the
through the interaction between the analyte and the imprecision to the negligible levels required for refer-
stationary phase contained in the chromatographic ence methods. The testosterone internal standard,
column, in the presence of a mobile phase flowing usually 13C2-testosterone or d3-testosterone, is added
in the system: a liquid solvent or a mixture of solvents at the very beginning of sample processing, and, since
in liquid chromatography, and a gas in gas chroma- the stable, isotopically labeled internal standard dis-
tography. The relative affinity between the analyte plays the same physical and chemical properties of the
and the mobile and stationary phase will determine analyte, it will display the same recovery of the analyte
the time needed by the former to reach the detector: in each step and the same efficiency of derivatization,
retention time is a specific feature of the analyte and is so that the ratio between the analyte and the internal
constant in stable chromatographic conditions; the standard remains constant throughout preparation
higher the affinity for the stationary phase compared and analysis. The internal standard differs from the
to the mobile phase, the longer the retention time. analyte in the m/z of the precursor ion and of the

63
Chapter 4: Measuring testosterone, DHT and SHBG

fragments, so that it can be specifically identified and treated samples by protein precipitation with acetoni-
quantified by the MS detector. In the quantification trile (Harwood and Handelsman 2009) or mixtures of
by isotopic dilution, the calibration curve is built with methanol/zinc-sulfate (Rauh et al. 2006; Ceglarek
the ratio between the signal of the analyte and the et al. 2009; Kushnir et al. 2011) followed by on-line
signal of the internal standard for each calibration purification on preparative columns mimicking
point (y-axis) against the nominal concentration the function of a SPE cartridge (Rauh et al. 2006;
(x-axis). Each sample is quantified through back cal- Guo et al. 2006): while steroids are retained on
culation of the analyte/internal standard ratio found the cartridge, salts and protein residuals are sent to
on the calibration curve. the waste. After clean-up, the sample is directed to the
chromatographic column for analysis.
4.3.1.2 Liquid chromatographytandem mass Reverse-phase C18 and C8 columns have been
spectrometry used for testosterone analysis, usually working with
In liquid chromatography the molecules are resolved water as polar phase and methanol or acetonitrile as
for their specific partition coefficient between a organic phases. The pH of the mobile phases can be
flowing mobile phase and the stationary phase. Since buffered by addition of formic acid, ammonium acet-
large amounts of liquids are continuously eluted at ate or ammonium formate. Compared to the other
high pressure, the combination with the high vacuum steroid hormones, testosterone displays a very good
system of the MS was hindered until the generation of revelation efficiency in positive ion mode. All soft
the atmospheric pressure ionization (API) source in ionization approaches have been tested: electrospray
the 1980s. This procedure is able to completely evap- ionization (ESI), atmospheric pressure chemical ion-
orate the liquid solvent and to transfer the charge to ization (APCI) and the latest generation of atmos-
the molecules without, or only minimally, altering pheric pressure photo-ionization (APPI) sources.
their structure: this soft ionization allows the gener- Ceglarek et al. (2009) compared the signal-to-noise
ation of the ionized molecule, or precursor ion. Liquid ratio (S/N) for several steroids with the three sources,
chromatography is a versatile technique: the liquid and ESI was reported to give the highest S/N for
environment allows, theoretically, the analysis of all testosterone, while Kushnir and colleagues (2006)
kinds of molecules, and, when combined with MS, reported higher S/N by APPI for the natural molecule
generally requires a reduced volume of sample and but higher S/N by ESI for the oxime derivative. Since
less demanding purification than gas chromatography. the performance of each source may vary between
The volume of serum required for testosterone instruments of the same type, it is difficult to establish
measurement largely depends on the ion source and a general rule. It has proved wise to validate the best
on sensitivity of the mass spectrometer detector, and S/N on all the sources available on ones own MS
may usually vary from 0.05 to 0.5 ml (Cawood et al. detector.
2005; Kushnir et al. 2011). The extraction approach The appellative tandem refers to triple quadru-
mostly used is LLE, and several examples were pole mass spectrometry in which the selection of the
reported using methyl tertiary butyl ether (MTBE) precursor ion is physically separated from the mol-
(Kushnir et al. 2006; Moal et al. 2007), diethylether ecule fragmentation. While in GC-MS the fingerprint
(Cawood et al. 2005; Yamashita et al. 2009) or ethyl of the molecule is generated at the same time as
acetatehexane mixtures (Harwood and Handelsman ionization and simultaneous fragmentation of the
2009; Fitzgerald et al. 2010). In order to achieve molecule, the API-MS/MS instruments allow the sep-
satisfying sensitivity using minimal sample volume, aration of the ionization and fragmentation steps. In
some groups used derivatization strategies generating multiple reaction monitoring (MRM) mode, the pre-
the testosterone oxime derivative, by incubating the cursor ion is filtered for its specific m/z by the first
extracted sample either with hydroxylamine solution quadrupole and fragmented in the second quadru-
(Kushnir et al. 2006; 2010), or the picolinic acid ester pole, which acts as a collision cell. The third quadru-
derivative (Yamashita et al. 2009). Derivatization pole filters the specific fragments generated by the
requires further purification to remove excess salts, analyte further by their m/z. The process is called
and on-line (Kushnir et al. 2010) or off-line (Kushnir transition, and for each analyte two transitions, each
et al. 2006; Yamashita et al. 2009) solid-phase extrac- targeting a different fragment, are monitored: one
tion (SPE) approaches have been used. Some groups for the quantification and one for the qualitative

64
Chapter 4: Measuring testosterone, DHT and SHBG

confirmation. Similarly to GC-MS, quantification by of 16 pmol/l (Chen et al. 2010). However, routine
isotopic dilution is recommended in LC-MS/MS, but, measurement of free testosterone will not become
in order to avoid reciprocal isotopic interference, feasible until faster and more simple procedures for
internal standards with the addition of 3 or more free-fraction separation are developed.
amu (M+3) should be chosen. Deuterated isotopes Interestingly, some groups reported the develop-
are cheaper and easily available, but 13C isotopes are ment of a method for the simultaneous detection of
more stable during sample extraction and ionization, both testosterone and DHT; the latter being a more
and should be preferred. challenging analyte for which satisfying sensitivities
One of the great advantages of LC-MS/MS is the were achieved through derivatization (Yamashita
possibility of measuring multiple analytes in the same et al. 2009) or with the latest generation of MS detect-
run. In multianalytical methods the choice of sample ors (Shiraishi et al. 2008; Harwood and Handelsman
volume, extraction, chromatographic separation and 2009; Kulle et al. 2010).
source should consider the requirements of each ana- In addition to high specificity and reliability,
lyte. In our own method (Fanelli et al. 2011), testos- another important aspect of LC-MS/MS technology
terone is measured together with eight other steroids is the low cost per sample, which should boost its
starting from 0.9 ml of serum, allowing good sensi- introduction to routine laboratories. If testosterone is
tivity for the whole panel. We adopted an SPE included in the multianalyte LC-MS/MS method,
approach followed by on-line purification in order approximately 5080% of the total costs can be saved
to lower the noise level, to extend the column life by using one LC-MS/MS run instead of individual
(after 5000 measurements the column performance immunoassays for each analyte. Further resources
is not changed) and the source cleanness during the can be saved considering the consequences of testoster-
measurement of a large number of samples, prevent- one immunoassay inefficiency, resulting in expenses
ing the decrease in sensitivity caused by the accumu- due to repeated measurements, re-examination of the
lation of contaminants. Due to the frequently patients, wrong therapy and unsatisfactory quality of
occurring isobaric pairs, a chromatographic gradient patient care. A present drawback of LC-MS/MS is the
of 12.5 min was used, followed by a washing step and requirement of highly experienced, ad-hoc personnel to
a re-equilibration step to ensure specificity of detec- deal with the analytical difficulties, the complexity of the
tion. In the case of testosterone, particular attention instruments and the possible pitfalls.
should be paid to its resolution from the epimer
epitestosterone, especially when short chromatog- 4.3.1.3 Comparative analytical performance
raphy is used. Moreover, in our hands the APCI and standardization
source provided good sensitivity for all analytes A number of comparative studies on the accuracy of
included in the panel. The most relevant publications testosterone measurement by different methods were
reporting the currently validated methods to measure reported in the last decade. For no other hormone
serum testosterone by LC-MS/MS are summarized in have the analytical methods ever been assessed,
Table 4.2. questioned and compared to such an extent as for
Methods for the direct measurement of free tes- testosterone. The poor reliability of immunoassays
tosterone were also proposed with both LC-MS/MS was systematically described in comparison to both
(Chen et al. 2010) and GC-MS (Van Uytfanghe et al. GC-MS and LC-MS/MS (Table 4.3)
2004). Both techniques can achieve satisfying sensitiv- In an early work in 1996, a direct chemilumin-
ity to measure free-fraction levels, but the main prob- escence competitive immunoassay was compared to
lem is obtaining the free fraction without altering the the GC-MS method developed by the authors,
equilibrium with the bound fraction. The free fraction reporting excellent accuracy and precision for male
was obtained by ultrafiltration, SPE purification, samples but high discrepancies, in terms of correl-
HPLC separation, derivatization and final injection ation and systematic bias, for female samples
in the GC-MS system, with a sensitivity of 1520 (Fitzgerald and Herold 1996). In 2003 Taieb et al.
pmol/l (Van Uytfanghe et al. 2004). In another extended the comparison to eight automated
approach, after ultrafiltration, the procedure is eased immunoassays and two direct radioimmunoassays
by LLE purification and subsequent injection in the (RIAs) among the most commonly used in worldwide
LC-MS/MS system, reaching a limit of quantification laboratories. The authors reported high inter-assay

65
Table 4.2 Main features of published LC-MS/MS methods for the measurement of circulating testosterone

Reference Sample Extractionb Derivatization Chromatography Total API MRM transition Internal Calibrator No. LLOQd
c
volume (ml); run time source standard matrix analytes
typea (min)
Quantifying Qualifying
Guo et al. 2004 760; S PP + on-line SPE No RP-C18 18 APPI 289\97 d2-T BSA 9 100 pg/ml
Cawood et al. 50; S or P LLE (diethyl ether) No RP-C18 4.75 ESI 289\97 d2-T Stripped S 1 0.3 nmol/l
2005
Guo et al. 2006 200; S PP + on-line SPE No RP-C8 11 APPI 289\109 d2-T BSA 12 n.d.
e e
Kushnir et al. 100; S LLE (MTBE) + SPE Hydroxylamine RP-C18 3 ESI 304\112 304\124 d3-T BSA 1 0.0345nmol/l
2006
Rauh et al. 100; S or P PP + on-line SPE No RP-C18 6 APCI 289\109 289\97 d5-T Saline 3 0.35 nmol/l
2006 solution
e e
Borrey et al. 200; S LLE (MTBE) + on- Hydroxylamine RP-C18 3 ESI 304\124 304\112 d3-T BSA 1 0.01 mg/l
2007 line SPE
Kalhorn et al. 100; S LLE (ethylacetate : Hydroxylamine RP-C18 7 ESI 304\124 d3-T H2O 2 500 amol/o.c.
2007 hexane = 1 : 4)
Moal et al. 500; S LLE (MTBE) No RP-C8 20 ESI 289\97 d5-T BSA 1 <0.17 nmol/l
2007
Tai et al. 2007 13 grams; S SPE + LLE (hexane) No RP-C18 62 ESI 289\97 d3-T MeOH 1 n.d.
Janzen et al. 15; S PP No RP-C18 6 ESI 289\109 d8-17OHP Steroid free 10 2.07 nM
2008 S
Licea-Perez 300; S LLE (MTBE) 2,3-pyridine- RP-C18 5 ESI 437\286 d3-T Stripped S 2 0.2 ng/ml
et al. 2008 dicarboxilic
anhydride
Shiraishi et al. 100; S LLE (ethylacetate : No Hypersil 16 ESI 289\109 d2-T Steroid free 2 0.069 nmol/l
2008 hexane = 3 : 2) S
Singh 2008 100; S or P PP + on-line SPE No RP-C18 7.5 APCI 289\97 289\109 d3-T n.d. 1 7 ng/dl
Turpeinen 250500; S LLE No RP-C18 10 ESI 289\97 d2-T MeOH 1 0.15 nmol/l
et al. 2008 (diethylether :
ethylacetate = 7 : 3)
Table 4.2 (cont.)

Ceglarek et al. 100; S PP + on-line SPE No RP-C18 4 APCI 289\97 d3-T Saline 9 0.05 mg/l
2009 solution
Chen et al. 250; S LLE No RP-C8 6.5 APCI 289\109 d5-T MeOH 1 0.056 nmol/l
2009 (MTBE
+ 80% MeOH :
heptane)
Harwood and 200; S LLE (ethylacetate : No RP-C8 >8.5 APPI 289\109 d3-T BSA 6 10 pg/ml
Handelsman hexane) + on-line
2009 SPE
Yamashita 200; S LLE (diethyl ether) Picolinic acid RP-C18 12 ESI 394\253 d3-T Stripped S 2 1 pg/0.2 ml
et al. 2009 + SPE
Fitzgerald et al. 1000; S or P LLE (ethylacetate : No RP-C18 7 ESI 289\109 289\97 d3-T H 2O 1 5 ng/dl
2010 hexane = 3 : 2)
Kulle et al. 100; P SPE No RP-C18 3.2 ESI 289\97 d8-17OHP Steroid free 3 3 ng/dl
2010 P
Kushnir et al. 200; S LLE (MTBE) + Hydroxylamine RP-C18 3.5 ESI 304\112 304\124 d3-T BSA 3 10 ng/l
2010 on-line SPE
e e
Salameh et al. 150; S or P On-line SPE No RP-C12 4.5 APCI 289\109 289\97 d5-T n.d. 1 0.3 ng/dl
2010
13
Fanelli et al. 900; S PP + off-line SPE + No RP-C8 21 APCI 289\97 289\109 C2-T BSA 9 0.019 ng/ml
2011 on-line SPE
Abbreviations: n.d., not determined; API, atmospheric pressure ionization; MRM, multiple reaction monitoring; LLOQ, lower limit of quantification; BSA, bovine serum albumin; MTBE, methyl tertiary butyl ether;
APCI, atmospheric pressure chemical ionization; APPI, atmospheric pressure photo-ionization; ESI, electrospray ionization; RP, reversed phase.
a
serum (S), plasma (P).
b
liquid-liquid extraction (LLE); protein precipitation (PP); solid-phase extraction (SPE).
c
testosterone (T), 17-hydroxyprogesterone (17OHP).
d
on column (o.c.).
e
quantitative or qualitative function not specified.
Table 4.3 Summary of the results of the main comparison studies between immunoassays and GC-MS (a) and LC-MS/MS (b) on samples from men (M), women (W) and children (C)

(a)
Reference Immunoassay Producer Sample n Regression Slope Intercept Correlation Mean difference
coefficient (RIA  LC-MS/MS
or %RIA/LC-MS/MS)
Fitzgerald and Herold ACS kit Ciba Corning Diagnostic W 44 Linear 0.72  GC-MS 1.21 nmol/l r2 0.31 0.89 nmol/l
1996 Corp
M 57 Linear 1.07  GC-MS 0.19 nmol/l r2 0.98 1.2 nmol/l
Taieb et al. 2003 Architect i2000 Abbott Laboratories W 54 Deming 1.64  GC-MS 0.20 nmol/l r 0.80 1.26 nmol/l
M 45 Deming 1.07  GC-MS 1.48 nmol/l r 0.95 0.21 nmol/l
ACS-180 Bayer Diagnostic W 55 Deming 1.63  GC-MS 0.53 nmol/l r 0.84 2.04 nmol/l
M 50 Deming 1.07  GC-MS 1.19 nmol/l r 0.95 2.53 nmol/l
Immuno-1 Bayer Diagnostic W 55 Deming 1.47  GC-MS 0.48 nmol/l r 0.68 0.56 nmol/l
M 50 Deming 1.10  GC-MS 1.48 nmol/l r 0.96 0.54 nmol/l
Vidas Bio-Mrieux W 51 Deming 1.16  GC-MS 0.58 nmol/l r 0.75 0.07 nmol/l
M 50 Deming 0.94  GC-MS 2.92 nmol/l r 0.92 4.67 nmol/l
Immulite 2000 Diagnostic Products W 53 Deming 2.57  GC-MS 0.82 nmol/l r 0.57 3.23 nmol/l
Corporation
M 50 Deming 0.79  GC-MS 1.80 nmol/l r 0.97 2.46 nmol/l
Vitros ECi Ortho-Clinical Diagnostics W 55 Deming 1.15  GC-MS 0.40 nmol/l r 0.76 0.05 nmol/l
M 50 Deming 0.94  GC-MS 2.03 nmol/l r 0.97 3.36 nmol/l
AutoDelfia Perkin-Elmer W 55 Deming 3.28  GC-MS 2.01 nmol/l r 0.78 2.68 nmol/l
M 50 Deming 0.95  GC-MS 1.98 nmol/l r 0.86 0.69 nmol/l
Elecsys 2010 Roche Diagnostic W 53 Deming 0.85  GC-MS 0.48 nmol/l r 0.72 0.74 nmol/l
M 50 Deming 0.93  GC-MS 3.02 nmol/l r 0.96 4.79 nmol/l
RIA Immunotech Beckman-Coulter W 54 Deming 1.43  GC-MS 0.46 nmol/l r 0.78 0.46 nmol/l
M 50 Deming 0.96  GC-MS 2.95 nmol/l r 0.95 4.19 nmol/l
Coat-A-Count DPC Dade Behring W 51 Deming 1.45  GC-MS 0.56 nmol/l r 0.89 0.49 nmol/l
M 50 Deming 1.06  GC-MS 1.36 nmol/l r 0.92 0.55 nmol/l
(b)
Wang et al. 2004 DPC coat-a-tube Diagnostic Products M 101 Deming 1.098  LC- 2.9 ng/dl ICC 0.968 9.70%
RIA Corporation MS/MS
HUMC-RIA In house; tracer and M 101 Deming 1.141  LC- 39.2 ng/dl ICC 0.948 9.70%
antibody by ICN MS/MS
Elecsys 2010 Roche Diagnostics M 101 Deming 1.167  LC- 75.5 ng/dl ICC 0.965 3.40%
MS/MS
Vitros ECi Ortho-Clinical Diagnostics M 101 Deming 1.233  LC- 118.4 ng/dl ICC 0.954 11.20%
MS/MS
Immulite 2000 Diagnostic Products M 101 Deming 0.881  LC- 28.6 ng/dl ICC 0.925 18.70%
Corporation MS/MS
ACS Centaur Bayer Diagnostic M 101 Deming 1.195  LC- 1.4 ng/dl ICC 0.919 15.90%
MS/MS
Guo et al. 2004 RIA ICN M and W 50 Lineara 0.919 0.064 nmol/l r 0.971
Pharmaceuticals
Cawood et al. 2005 Extraction RIA In house W 3 nmol/ 67 n.d. 0.69  RIA 0.15 nmol/l r2 0.66
l
M 3 nmol/ 29 n.d. 0.74  RIA 0.26 nmol/l r2 0.35
l
W >8 nmol/ 13 n.d. 1.12  RIA 1.7 nmol/l r2 0.77
l
M >8 nmol/ n.d. n.d. 1.02  RIA 0.2 nmol/l r2 0.97
l
Kushnir et al. 2006 Vitros ECi Ortho-Clinical Diagnostics M and W 216 Deming 1.15  LC- 11.2 ng/dl r 0.976
MS/MS
<50 ng/dl 150 4.1 ng/dl
Rauh et al. 2006 Roche Elecsys Roche Diagnostics C (<5 nmol/ 70 Passing- 1.485  LC- 0.149 nmol/l r 0.623
l) Bablok MS/MS
C (>5 nmol/ 37 0.954  LC- 0.142 nmol/l r 0.864
l) MS/MS
Borrey et al. 2007 RIA Spectria Orion Diagnostica W 43 Passing- 0.887  RIA 0.014 mg/l r2 0.92 0.031 mg/l
Bablok
Moal et al. 2007 CIS bio Shering SA W and C 59 Deming 0.972  LC- 0.3 nmol/l r 0.870 0.23 nmol/l
MS/MS
Immunotech Beckman-Coulter W and C 59 Deming 1.365  LC- 0.8 nmol/l r 0.868 1.24 nmol/l
MS/MS
Spectria-Orion Orion Diagnostica W and C 59 Deming 1.177  LC- 0.2 nmol/l r 0.803 0.38 nmol/l
MS/MS
Liaison DiaSorin W and C 59 Deming 1.114  LC- 0.7 nmol/l r 0.772 0.81 nmol/l
MS/MS
Roche Modular E Roche Diagnostics W and C 59 Deming 1.153  LC- 0.1 nmol/l r 0.792 0.32 nmol/l
MS/MS
Ceglarek et al. 2009 Elecsys 2010 Roche Diagnostics n.d. 57 Passing- 1.083  LC- 0.089 mg/l Pearsons 0.967 9.70%
Bablok MS/MS r
Shiraishi et al. 2008 Coat-A-Count DPC Diagnostic Products M and W 145 Deming 1.06  RIA 20.6 nmol/l r2 0.9785 0.15 nmol/l
Corporation
Singh 2008 Direct ACS:180 Bayer M 37 n.d. 1.0497  RIA 8.95 ng/dl r2 0.9809
Extraction ACS:180 Bayer W 73 n.d. 0.7231  RIA 0.2109 ng/dl r2 0.8561
Turpeinen et al. 2008 Spectria coated Orion Diagnostica >3 nmol/l 121 Linear 0.57  LC- 2.3 nmol/l r 0.92 43%
tube RIA MS/MS
Extraction RIA In house >3 nmol/l 109 Linear 0.85  LC- 0.18 nmol/l r 0.984 15%
MS/MS
Chen et al. 2009 Abbott Architect Abbott Laboratories >3 nmol/l <62 n.d. 0.8710  LC- 1.4450 nmol/l r2 0.9406
i2000 MS/MS
<3 nmol/l <62 n.d. 0.9434  LC- 1.0093 nmol/l r2 0.3408
MS/MS
Roche Modular Roche Diagnostics >3 nmol/l <62 n.d. 0.8898  LC- 0.4237 nmol/l r2 0.9483
E170 MS/MS
<3 nmol/l <62 n.d. 0.9657  LC- 0.551 nmol/l r2 0.3836
MS/MS
DPC Immulite Diagnostic Products >3 nmol/l <62 n.d. 0.7602  LC- 1.6192 nmol/l r2 0.8124
2500 Corporation MS/MS
<3 nmol/l <62 n.d. 0.8623  LC- 0.5480 nmol/l r2 0.2925
MS/MS
Harwood and RIA Delfia Perkin Elmer M 172 Passing- 1.333  LC- 0.35 ng/ml r 0.9631 42%
Handelsman 2009 Bablok MS/MS
Salameh et al. 2010 Extraction + In house <40 ng/dl n.d. n.d. 1.0064  RIA 0.0618 ng/dl r2 0.995
LC RIA
Entire range n.d. n.d. 1.0210  RIA 1.4997 ng/dl r2 0.994
Brandhorst et al. 2011 Elecsys Roche Diagnostics M 117 Passing- 1.06  LC- 0.02 mg/l Pearsons 0.974
Testosterone Bablok MS/MS r
W 119 Passing- 1.32  LC- 0.03 mg/l Pearsons 0.893
Bablok MS/MS r
Elecsys Roche Diagnostics M 117 Passing- 1.05  LC- 0.03 mg/l Pearsons 0.986
Testosterone II Bablok MS/MS r
W 113 Passing- 1.06  LC- 0.02 mg/l Pearsons 0.956
Bablok MS/MS r
Fanelli et al. 2011 Elecsys Modular Roche Diagnostics M 51 Deming 1.004  LC- 0.097 ng/ml r 0.938 2.50%
E170 MS/MS
W 111 Deming 1.724  LC- 0.181 ng/ml r 0.773 16.80%
MS/MS
Abbreviations: n.d., not determined; RIA, radioimmunoassay; ICC, interclass correlation coefficient; n, number of tested samples.
a
x-axis and y-axis method not specified.
Chapter 4: Measuring testosterone, DHT and SHBG

variability among the 10 immunoassays, and, through apply their own LC-MS/MS technique, raising the
the comparison with a GC-MS method, concluded problem of harmonization, validation, calibration,
that none of them showed acceptable reliability for quality control material and, finally, reference inter-
the measurement of female or pediatric testosterone vals using all these different LC-MS/MS methods
levels. Furthermore, two methods were reported to (Rosner et al. 2010).
have questionable reliability even in the male range In the last few years, papers have reported the
(Taieb et al. 2003). In 2004 Wang et al. published a comparison to GC-MS reference methods of several
similar comparative study between four automated LC-MS/MS methods, using different preparative
immunoassays, two RIAs and LC-MS/MS on male methodologies, chromatography and detection strat-
serum. The authors found that only four methods egies (Thienpont et al. 2008; Vesper et al. 2009).
gave results within 20% of those obtained by LC- Thienpont et al. (2008) reported the comparison of
MS/MS in more than 60% of samples. In addition, four routinely used LC-MS/MS methods with a refer-
when samples with testosterone concentrations below ence GC-MS method listed by the Joint Committee
100 ng/dl were considered, all six immunoassays for Traceability in Laboratory Medicine (JCTLM).
failed to give results with acceptable variability in The LC-MS/MS methods reported good intra-labora-
55.5 to 90.9% of the samples. The authors concluded tory precision in the entire range of clinically relevant
that none of the automated immunoassays displayed values: the median coefficient of variation (CV) was
adequate precision, accuracy and sensitivity to be below 6.6% at concentrations > 5 nmol/l, and below
applied to samples from females and children (Wang 8.5% at concentrations < 5 nmol/l. The regression
et al. 2004). analysis, in terms of slope and intercept coefficients,
Moal et al. (2007) reported another multi-assay the correlation coefficients and the percentage of the
comparison where one extractive RIA, two direct difference plot, revealed good agreement of the four
RIAs and two automated immunometric platforms LC-MS/MS methods compared to the reference GC-
were compared to a validated LC-MS/MS method MS method: far better than that obtained by similar
on female and pediatric sera. Confirming previous studies in which immunoassays were compared to
findings, they reported poor agreement between the GC-MS (Taieb et al. 2003). The authors stated that
immunoassays and LC-MS/MS, in terms of both poor this improvement in performance should be ascribed
correlation coefficient and variable slope and inter- to the use of isotope dilution and to the specificity
cept coefficient by regression analysis. In general, all provided by the precursor-to-fragment transitions
methods overestimated the results. The authors performed by tandem MS. The authors concluded
attributed this problem not only to the poor specifi- that the tested LC-MS/MS procedures displayed suffi-
city of the immunoassays, but also to non-linearity of cient levels of sensitivity in defining normogonadal,
the calibration curves, especially at low doses, where hypogonadal and female testosterone levels, proper
the low-concentration samples appear and high vari- specificity and good accuracy. However, small to
ance is expected (Moal et al. 2007). medium biases (<9.6%) were reported for three out
The poor performance of immunoassays at low of four assays (Thienpont et al. 2008).
concentrations was confirmed by Cawood et al. Vesper et al. reported the results of the compari-
(2005), who verified the accuracy of their LC-MS/ son of seven LC-MS/MS methods and one GC-MS/
MS method with two GC-MS methods. Compared MS method to an LC-MS/MS reference method per-
to the LC-MS/MS method, an in-house extractive formed at the National Institute of Standard and
RIA showed good correlation and regression for levels Technology (NIST) included in the JCTLM list
above 8 nmol/l, but poor correlation and regression at (Vesper et al. 2009). The reported intra-laboratory
levels below 3 nmol/l. CV ranged between 2.52 and 25.58% at low levels of
Many other papers reported similar results on the testosterone (8.47 ng/dl) and between 1.40 and
comparison of LC-MS/MS technique and one or 11.36% at high levels of testosterone (297 ng/dl).
more immunoassays of different kinds (Table 4.3). The reported inter-laboratory CV is < 15% at concen-
All these efforts provided strong and unambiguous trations above 44 ng/dl, but it increased up to 33% at
evidence for the need to shift toward a more specific, lower levels. The percentage difference between each
sensitive and reliable method such as LC-MS/MS. As tested procedure and the reference method is on
a consequence many laboratories all over the world average 7.5% for concentrations > 100 ng/dl and

72
Chapter 4: Measuring testosterone, DHT and SHBG

15.5% for concentrations < 100 ng/dl. By performing improved, the overall performance of current
a regression analysis, proportional bias was observed immunoassays is not much different from that of 40
in six assays, while constant bias was observed in four years ago. For instance, the early assays already
assays. The authors stated that the variability showed a lower detection limit of 310 pg, as do
observed among the tested tandem-MS procedures is current assays. Since RIAs are most sensitive at low
surprisingly small for testosterone levels above antibody concentrations, when competition between
100 ng/dl; better than what was previously reported tracer and unknown is high, high-affinity antibodies
for immunoassays. However, the situation worsens at are crucial for sensitive assays. However, usually high
female concentrations, where the increased variability affinity is better obtained with polyclonal antisera
observed could be attributed to several factors: the which display elevated cross-reactivity with DHT.
sensitivity limit of the different assays, intra-labora- A substantial improvement in the sensitivity of tes-
tory variability, interference in the detection (ion sup- tosterone RIAs (and other immunoassays as well)
pression) caused, probably, by phospholipids, and the could be achieved by using highly specific monoclo-
rounding to different decimal points. When com- nal antibodies with high affinity: a goal very difficult
pared directly to the NIST method, the single assays to reach.
reported a mean difference below or equal to 10%, In general the current in-house methods for tes-
which is smaller than that reported for immunoassays tosterone RIA are the same as the early assays of the
(Vesper et al. 2009). seventies and are still in use mainly for research
Both papers concluded by emphasizing the purposes because they are cheap and accurate. An
importance of careful validation and standardization extraction step is necessary to eliminate serum pro-
of the procedure and of monitoring performance in teins which do not allow the correct interaction of
each laboratory, together with the importance of albumin and SHBG-bound testosterone with the
proper assay calibration and quality control. Finally, antiserum.
they promote the sharing of common calibration The long experience with the traditional RIAs
materials and participation in external quality assur- produced well-documented reference intervals in dif-
ance programs and/or the comparison with inter- ferent age and sex groups, and this methodology has
nationally accepted reference methods (Thienpont several advantages such as high sensitivity, due to the
et al. 2008; Vesper et al. 2009). large volume of samples which can be employed, and
the possibility of measuring different steroids in the
same sample extract. In addition, the extract can be
4.3.2 Radioimmunoassay subjected to chromatography, allowing the separation
The first radioimmunoassay for plasma testosterone and measurement of different steroids, for example
was developed at the end of the sixties (Furuyama DHT, androstenedione, etc.
et al. 1970). It was based on an antiserum raised Some disadvantages should be listed as well. First,
against testosterone coupled to bovine serum albumin without doubt it is a challenging, time-consuming
at position 3 (T-3-BSA), 3H-labelled testosterone as and costly assay and it requires high technical compe-
the tracer and bound/free separation by ammonium tence and accurate monitoring of the extraction effi-
sulfate precipitation. Plasma testosterone was ciency. Sometimes it may be difficult to obtain a large
extracted and chromatographed on alumina columns volume of sample, e.g. from children, and the RIA
prior to immunoassay. Radio- and other immuno- may be affected by matrix effect (matrix difference
assays for plasma and serum testosterone were between serum samples, particularly hemolyzed and
developed by several investigators, for instance Nie- lipemic samples, and solution of standards used to
schlag and Loriaux (1972). These authors produced prepare the standard curve) and by the presence of
their own antiserum by a novel immunization tech- auto-antibodies leading to falsely high or low values,
nique (Vaitukaitis et al. 1971) and distributed the depending on the type of antibody interaction occur-
antiserum freely to other laboratories so that their ring. Finally, we should not forget the problems
method became widely used and their papers were related to organic solvents and radioactive waste dis-
ranked as Citation Classics in 1981. Slowly, kit manu- posal and to safety concerns.
facturers took over the development of assays, but Because of these problems, traditional RIAs have
while the practicability of the assays consistently been almost completely replaced by non-extraction

73
Chapter 4: Measuring testosterone, DHT and SHBG

methods for clinical use. These kinds of commercially binding to the antiserum. The primary antibody
available RIAs are either based on double antibody can be poly- or monoclonal and is usually bound
separation or are in solid phase; i.e. the antibody is to the tube wall or to the plate well. In EIAs/
fixed at the wall of the reaction tubes (coated tubes) so ELISAs the tracer is represented by testosterone
that no centrifugation is required, reducing the coupled to an enzyme (alkaline phosphatase, b-
hands-on time and improving practicability. Serum galactosidase, penicillinase, acetylcholinesterase or
testosterone is displaced from carrier proteins by horse radish peroxidase) which starts a colorimetric
chemical agents competing for protein binding, e.g. reaction upon addition of the substrate at the end of
danazol, although the exact nature of the kit compon- the incubation time. This results in color develop-
ents is usually known only to the manufacturers and ment which is inversely proportional to the amount
is protected by property rights. Well-validated non- of unlabelled testosterone and can be read by a
extraction methods may work well for male serum spectrophotometer. The sensitivity of EIA/ELISA is
samples; although inaccurate testosterone concentra- affected by the number of steroid molecules coupled
tions are occasionally measured in individual samples to the enzyme, and the proper molar ratio between
containing abnormal SHBG concentrations or sub- steroid and enzyme must be carefully validated (Ras-
stances (e.g. drugs) interfering with the kit compon- saie et al. 1992). In FIA the tracer is testosterone
ents. However, results obtained with different RIA coupled to a molecule (e.g. europium) which fluor-
kits have been repeatedly reported to be poorly com- esces upon stimulation. The fluorescence is measured
parable (Jockenhvel et al. 1992; Boots et al. 1998). by a fluorimeter and, again, is inversely related to the
There are a number of advantages of the direct amount of cold testosterone contained in the sample.
RIA methodology. First of all it is relatively simple to Critical in EIAs/ELISAs and FIAs are the washing
carry out, it requires a small volume of samples (usu- steps, which should be carefully carried out in order
ally 0.1 ml or less) and it can be automated, resulting to eliminate non-specifically bound substances which
in a very fast turnaround time, rendering it particu- would result in poor precision and falsely elevated
larly suitable to the large number of samples tested in readouts.
a routine clinical laboratory. It is also relatively inex- Automatic multianalyzers, mainly based on non-
pensive and generally fairly accurate for levels above radioactive methods requiring a low sample volume
300 ng/dl (healthy men). There is some evidence that (usually less than 0.1 ml) are now available and widely
a well chosen and performed RIA may offer equal used in diagnostic clinical laboratories for the direct
precision to LC-MS assay (Legro et al. 2010). There and quick measurement of serum testosterone. Some
are, however, downsides to direct RIAs: concentra- of these systems have been evaluated against the ref-
tions of testosterone can be often overestimated, espe- erence method based on MS, showing acceptable
cially when levels sink below 10 nmol/l. Moreover, results at least in male samples (Fitzgerald and Herold
they are poorly standardized, reference ranges vary 1996; Levesque et al. 1998; Gonzalez-Sagrado et al.
between assays, and direct RIAs share with extractive 2000). However, inconsistency of results obtained
RIAs the same problems of matrix effect, radioprotec- with different methods is reported as well, and some
tion and radioactive waste disposal. systems seem to suffer from systematic problems,
resulting in over- or underestimation of serum testos-
terone and/or insufficient sensitivity, especially in
4.3.3 Other immunoassays female samples (Taieb et al. 2002). This is very evi-
The most popular alternatives to radioactive dent when comparing the results of external quality
methods are immunoassays based on non-radio- control trials (Middle 2002).
actively labeled tracers such as fluoroimmunoassay The quality control program of the College of
(FIA), chemiluminescent assay (CLIA) and enzyme- American Pathologists clearly demonstrates how
linked immunosorbent assay (EIA/ELISA). They can great the inaccuracy of total testosterone measure-
be in liquid or in solid phase, whereby solid phase, ment is, even though one-third of the clinical labora-
microtiter plate-based assays are preferred due to tories use the same automated platform and three of
easy handling and proneness to automation. Also in these platforms cover two-thirds of the market (Ros-
these assays it is the antigen that is labeled and ner et al. 2007). The comparison of results obtained
competes with the endogenous testosterone for by immunoassays with those derived from MS shows

74
Chapter 4: Measuring testosterone, DHT and SHBG

a variability in testosterone levels between 4.7 and 4.3.4 Assessment of free testosterone
2.6 nmol/l in adult males and between 0.7 and 3.3
The direct measurement of free testosterone in serum
nmol/l in females (Taieb et al. 2003). In such condi-
is based on the same principles governing the assay of
tions, it is sometimes difficult to distinguish hypo-
free thyroid hormones, and has been extensively con-
and eugonadal males, unless the laboratory has opti-
sidered and reviewed by R. Ekins in the past (Ekins
mized its own method and population-related refer-
1990). Serum testosterone exists in an equilibrium
ence intervals. In any case, the diagnosis of
between free and protein-bound fractions: an equilib-
hypogonadism requires multiple determinations of
rium which is invariably disturbed by all methods of
testosterone levels during 24 hours, with the inherent
free-hormone measurement; a factor that should be
costs.
kept in mind when choosing a method and analyzing
Furthermore, it seems impossible to trust the
the data. The methods of reference for free-hormone
testosterone levels detected in women and children
analysis are equilibrium dialysis and ultrafiltration.
with these immunoassays (Wang et al. 2004). When
womens samples are processed, all automated
immunoassays seem to be affected by the interfer- 4.3.4.1 Equilibrium dialysis
ence of other steroids, such as dehydroepiandros- In equilibrium dialysis, the serum (dialysand) is put
terone sulfate (DHEAS) (Warner et al. 2006; Middle in contact with a buffer (dialysate) through a mem-
2007), and of drugs, such as danazol and brane which allows the passage of low-molecular-
mifepristone (RU486), thus introducing more vari- weight compounds (e.g. free hormones) but retains
ability and difficulty in result interpretation. This the binding proteins. As a consequence of the passage
lack of specificity seriously affects clinical manage- of free hormone molecules to the dialysate, new hor-
ment in the newborn period and early infancy, mone molecules will dissociate from the binding pro-
where poor clinical correlation to steroid test levels teins until a new equilibrium is reached and the free-
is very frequent and sensitivity is not adequate hormone concentration is the same on the two sides
enough to differentiate prepubertal and pubertal of the membrane. The free hormone can now be
secretion. measured in the dialysate either directly (e.g. by
A practical approach, as suggested by Kane et al. RIA) or indirectly by knowing the total hormone
2007), is to adopt a commercial total testosterone concentration and assessing the percentage of added
assay only if it provides validation against an MS labeled hormone passed in the free fraction. Provided
assay and the regression plot of the data obtained by that the ion composition of the buffer does not inter-
the two methods is supplied. The slope would give fere with the equilibrium constant (K), dialysis is
information about its accuracy and the intercept thermodynamically equivalent to serum dilution and
about its reliability at low concentrations. It may also leads to a reduction of the free-hormone concentra-
be a good laboratory strategy to analyze male and tion and to dissociation of new hormone from the
female samples for total testosterone with two differ- binding proteins until a new equilibrium is estab-
ent calibrated assays. lished in the system. At equilibrium the original
As for every other method, each laboratory free-hormone concentration is therefore only
should carefully validate the results obtained by the approximately maintained because, since the total
multianalyzer before it is implemented for routine hormone concentration is constant, the final, meas-
testosterone measurement. In practice, however, val- ured free-hormone concentration will be somewhat
idation is limited by the fact that most of the systems diluted and lower than that in the original sample.
are based on a master calibration curve carried out This effect can be regarded as negligible if the relative
by the manufacturer and not available to customers. free-hormone concentration is low, as in the case of
Each assay then requires only one or two calibrators free thyroxin (0.02%), but might become relevant in
to adjust the master curve and parallelism tests the case of free testosterone (2%), so that the buffer
cannot be performed. As in the non-automatic volume against which the sample is dialyzed should
assays, differences observed between the kits can be be kept to a minimum. In this respect, it is the total
ascribed to differences in the matrix of the calibra- volume of dialysand plus dialysate which determines
tors and in the affinity, titer and specificity of the the dilution factor; while the position of the dialysis
antibodies used. membrane between the two compartments, i.e. the

75
Chapter 4: Measuring testosterone, DHT and SHBG

individual volume of the two compartments, is irrele- approach compared to the more common methods
vant for the free-hormone concentration, which, at based on analog free testosterone measurement or
equilibrium, will be the same on the two sides (Ekins calculated free testosterone (Chen et al. 2010).
1990).
High technical competence is required for this 4.3.4.3 Direct free testosterone assays
methodology and represents an important limitation. The principles of the direct free testosterone methods
Each assay step introduces a specific variability source have been described in detail in the previous edition
that contributes to the total variability and increases of this book (Simoni 2004). In practice such assays are
the risk of error. The purity of the tracer is crucial, as based on a labeled compound to be added to the
the presence of a contaminant that does not link to sample, the analog, which is totally non-reactive
proteins can enhance the concentration of detected with serum proteins, but can be recognized by the
free testosterone. Finally, accuracy is affected by the solid phase antibody present in a limited amount and
accuracy of the total testosterone assay used. which competes for binding with the free hormone in
the sample.
4.3.4.2 Ultrafiltration Several commercially available kits are based on
The problem of sample dilution is avoided in the case this principle and are widely used to assess free tes-
of ultrafiltration of undiluted serum, the second ref- tosterone in clinical samples. Unfortunately, there is
erence method in free-hormone determination. In evidence that these methods are totally inaccurate
this procedure a serum sample is centrifuged through (Fritz et al. 2008; Chen et al. 2010). In fact, the direct
a membrane with an appropriate molecular-weight measurement of free testosterone in serum based on
cutoff. Only free hormone and low-molecular-weight the labeled hormone analog is valid only if the
compounds will be collected in the ultrafiltrate at a analog does not interact with the serum proteins: a
concentration equal to that in the original sample. condition which is currently not met by commercial
The free hormone can be directly assessed by RIA of kits. In fact, neither the identity of the analog tracer,
the ultrafiltrate or by indirect measurement of the nor the validation of the kit (showing the absence of
relative fraction of labeled hormone added to the interactions with the serum protein) is usually dis-
original samples which is recovered in the ultrafiltrate closed by the manufacturer. On the contrary, the
(Vlahos et al. 1982). Direct measurement by RIA is analog principle is often not even mentioned or is
preferable both in ultrafiltration and in equilibrium misrepresented in the instructions accompanying the
dialysis, since the impurities of the tracer can result in kits, which are often validated only against other kits
inaccurate estimation of the free fraction. Ultrafiltra- and not against dialysis or ultrafiltration. It should be
tion devices are commercially available (e.g. Centri- kept in mind that, in practice, finding a hormone
free micropartition system, Millipore). analog totally unreactive with serum proteins is very
Possibly ultrafiltration may be limited if non-fil- difficult, and several studies have shown that such an
terable binding competitors are present in the sample, interaction indeed occurs, resulting in inaccurate
or if binding proteins interact with the membrane; measurements of free testosterone. In this respect it
this will result in progressive increase of the free- is interesting that serum free testosterone measured
hormone concentration in the filtrate. In addition it by an analog method accounts for 0.50.65% of
is affected by temperature, so that a strict control of total testosterone, while equilibrium dialysis and
this parameter is necessary. However, since the ultra- ultrafiltration give values of 1.54%, revealing incon-
filtration time is rather short (one hour or less) com- sistencies between the different approaches (Rosner
pared to dialysis (several hours), the possible changes 1997; Winters et al. 1998). In a direct comparison,
in the equilibrium ensuing from these and other free testosterone values measured by a bestseller
factors may be assumed to be negligible. As for equi- analog kit were only 2030% of those measured by
librium dialysis, the testosterone RIAs accuracy dir- equilibrium dialysis (Vermeulen et al. 1999).
ectly affects the accuracy of the methodology which is For these reasons the kits for direct free testoster-
not trustable at low concentrations. one measurement presently available do not measure
Recently, free testosterone has been measured by what they claim and should not be used (Rosner 2001;
LC-MS/MS following ultrafiltration or equilibrium Swerdloff and Wang 2008). If LC-MS/MS coupled to
dialysis, demonstrating the superiority of such an ultrafiltration becomes technically affordable, this

76
Chapter 4: Measuring testosterone, DHT and SHBG

approach is likely to become the method of choice for measurement in saliva, several authors attempted
assessment of free testosterone in clinical serum LC-MS/MS of salivary testosterone with very promis-
samples. ing results (Matsui et al. 2009; Macdonald et al. 2011).
With the diffusion of MS-based methodologies, this
4.3.4.4 Salivary testosterone approach might offer an accurate and straightforward
Salivary testosterone is considered to be a good index assessment of free testosterone in the near future.
of serum free testosterone, and a highly significant
correlation with serum total and free testosterone has 4.3.4.5 Bioavailable testosterone
long been known (Wang et al. 1981). Recently a Several lines of evidence suggest that not only free
strong correlation with bioavailable testosterone has testosterone but also albumin-bound testosterone is
been demonstrated (Morley et al. 2006). As with other available to the target tissues for biological activity
steroid hormones, free testosterone enters saliva (Manni et al. 1985). Therefore, the non-SHBG-bound
though passive diffusion across the acinar cells of testosterone is called bioavailable testosterone. This
the salivary glands and can be measured directly in parameter can be measured based on the property of
saliva by RIA or EIA with or without extraction. ammonium sulfate to precipitate SHBG together with
The advantages and disadvantages of saliva as a the steroids bound to it. Trace amounts of labeled
diagnostic medium have been extensively reviewed testosterone are added to the samples and, after
(Lewis 2006; Levine et al. 2007; Chiappin et al. allowing for equilibration with endogenous testoster-
2007). Based on a non-invasive, stress-free and easily one, an equal volume of a saturated solution of
repeatable sampling technique even outside the clin- ammonium sulfate is added (final concentration
ical setting, the assay is well suited for pediatric appli- 50%) and SHBG is separated by centrifugation. The
cations, for screening male hypogonadism (Morley percentage of labeled testosterone remaining in the
et al. 2006) and for investigating biosocial models supernatant represents an estimate of bioavailable
and testosteronebehavior relationship (Dabbs 1990; testosterone, which can be calculated knowing the
Shirtcliff et al. 2001; 2002; Booth et al. 2003; Granger total testosterone concentration of the sample (Manni
et al. 2004). et al. 1985).
The manner in which saliva samples are collected Alternatively, testosterone can be measured dir-
and stored may influence testosterone measurement, ectly in the supernatant by RIA (Dechaud et al. 1989).
more than other analytes such as cortisol and DHEA, The ammonium sulfate concentration is critical to
because of substances that can be used to stimulate proper precipitation of SHBG only, and this param-
saliva flow, the presence of a transient rise in testos- eter should be accurately tested in each laboratory in
terone levels in the first few minutes after chewing the order to avoid precipitation of albumin as well, and
gum, and the presence of blood contamination from underestimation of bioavailable testosterone (Davies
oral microinjuries (Granger et al. 2004; Atkinson et al. 2002). In spite of the apparent simplicity of the
et al. 2008). Storage temperature and time are two assay, relevant technical differences in the method-
variables that dramatically influence salivary testos- ology and in the algorithms applied in various labora-
terone assays (Granger et al. 2004). The study of the tories are the cause of often inaccurate results (De
effect of five different clean-up methods on salivary Ronde et al. 2006). A possible way to circumvent this
steroid and protein concentration demonstrated that problem is represented by the immunocapture of
testosterone and DHEA were the analytes more SHBG and SHBG-testosterone complexes by mono-
strongly affected by pretreatment methods, calling clonal antibodies coupled to magnetic beads, followed
for a careful validation of saliva sampling procedures by magnetic separation and measurement of non-
before their adoption in clinical laboratory routine SHBG-bound testosterone by RIA in the supernatant
(Atkinson et al. 2008). (Raverot et al. 2010).
Since salivary testosterone provides a measure of When properly done, bioavailable testosterone
free testosterone, its assessment has been considered correlates quite well with total testosterone and calcu-
especially in those conditions in which an elevation of lated free testosterone. However, it is unclear, at the
serum concentrations of SHBG can hinder the accur- moment, whether the estimation of bioavailable tes-
ate diagnosis of hypogonadism, for example in male tosterone provides any superior diagnostic power
aging. Given the problems related to direct over the accurate assessment of the latter parameters

77
Chapter 4: Measuring testosterone, DHT and SHBG

in clinical conditions of mild hypogonadism accom- bioavailable testosterone and other androgens, several
panied by increased SHBG and possibly reduced con- in-vitro androgen bioassays have been developed.
centrations of serum albumin, such as in LOH. Currently six cell-based androgenic bioassays, based
on detection of intracellular, AR-dependent events,
4.3.4.6 Calculated free testosterone have been reported in the literature (Roy et al. 2008;
Assessment of free testosterone is recommended by Need et al. 2010 and references therein). Such bio-
several national and international scientific societies assays are interesting especially for screening and
in consensus guidelines for the evaluation of andro- detecting substances with androgenic activity; for
gen deficiency when serum total testosterone levels example endocrine disruptors (Soto et al. 2006). They
are borderline. The classical methods for free and are usually based on reporter genes (e.g. luciferase),
bioavailable testosterone measurement reported stimulated by the activation of the AR induced by the
above (i.e. equilibrium dialysis, ultrafiltration and testosterone/androgens present in the test sample.
ammonium sulfate precipitation), however, are too Both permanent and transient transfection systems
cumbersome for clinical routine. This is the main have been tried.
reason why analog methods became so widespread These bioassays have a sensitivity of about 0.81
despite their unreliability. A very popular alternative nM testosterone equivalents and are specific for
is provided by the calculation of free testosterone androgens, but it is unclear which portion of circulat-
starting from measured concentrations of total testos- ing testosterone they actually measure. In fact, when
terone and SHBG and based on equilibrium-binding long incubation times are used, testosterone dissoci-
theory or empirical equations. There are at least five ates from SHBG, affecting the equilibrium between
published formulae for free testosterone calculation, bound and free/biologically available fraction. Other
some of them available online (e.g. www.issam.ch/ factors influencing the neat, end effect are the influx
freetesto.htm), even as downloadable applications of the hormone into the cell and the intracellular
for tablets and smart phones. metabolism. In addition, bioassays do not consider
These formulae have been recently reviewed and the passage of the hormone from the capillary blood
validated for their accuracy against equilibrium dialy- flow into the tissues/cells. Inevitably every bioassay
sis in over 2000 serum samples. This study demon- represents a static system, as opposed to the very
strated that commonly used formulae systematically dynamic, in-vivo situation. Long-term incubation
overestimate free testosterone compared to laboratory bioassays correlate better with free/bioavailable tes-
measurement (Ly et al. 2010). Interestingly, calcula- tosterone; while short-term incubation bioassays cor-
tions based on equilibrium-binding equation relate better with total testosterone. Therefore, the
methods performed even worse than assumption-free, question arises of whether bioassays are really able
empirical formulae, revealing that the equilibrium- to measure a unique, physiologically relevant fraction
binding approach relies on only partially correct of serum testosterone which cannot be detected and
hypotheses. For instance, the reported value of the quantified by other methods and could be useful in a
TestosteroneSHBG affinity constant differs by up diagnostic setting. For the time being, this does not
to fourfold between different authors; possibly seem to be the case (Need et al. 2010).
because it is still not clear if this constant is age
related. Therefore, if calculated free testosterone is 4.4 Measurement of
used in clinical practice, the physician must be aware
of the limitations of such an approach and should not
dihydrotestosterone
draw therapeutic decisions based only on this The principles and the methods for testosterone assay
parameter. are basically valid for DHT as well. The main problem
in DHT measurement is the elevated cross-reactivity
of all polyclonal antisera with testosterone, which
4.3.5 Bioassay renders the direct, accurate quantification of DHT
Immunological methods measure just the mass of in a male serum sample impossible. As a conse-
circulating testosterone, not its bioactivity. Under quence, DHT has to be separated from testosterone
the assumption that the overall androgenic bioactivity before measurement or, alternatively, testosterone
in serum and peripheral tissues results from must be chemically modified so that it is not

78
Chapter 4: Measuring testosterone, DHT and SHBG

recognized by the antibody. At present there are only 4.5.1 Choice of the kit and assay validation
two ways of quantifying DHT in serum accurately:
Presently most laboratories choose to measure
after chromatographic separation or after oxidation
serum testosterone and DHT by commercially
of testosterone. Both methods involve an extraction
available kits. In all cases the final adoption of a
step.
kit for routine determination should depend on an
The classical methods for chromatographic separ-
in-house re-validation of the candidate assay. This
ation of DHT and testosterone are HPLC and celite
re-validation includes an assessment of the sensitiv-
chromatography, described in detail in the previous
ity, specificity, accuracy and intra-assay precision of
edition of this chapter (Simoni 2004). Recently, sev-
the kit. The specificity and the accuracy of the assay
eral authors applied GC-MS or LC-MS/MS to the
are checked by performing cross-reactivity, parallel-
measurement of DHT, and the simultaneous meas-
ism and recovery tests. These tests are usually
urement of testosterone and DHT without derivatiza-
carried out by the kit manufacturer beforehand
tion in clinical samples has been validated (Shiraishi
and the data are reported in the kit instructions,
et al. 2008). This approach allows rapid, specific and
but it is good practice to repeat them in ones own
accurate measurement, and it is conceivable that MS
laboratory, since experience shows that many
measurement of DHT will gain more popularity in
systems fail to deliver what they promise. Parallel-
the near future.
ism and recovery tests are crucial and should be
Several direct DHT kits using microtiter plate-
performed with several different individual serum
based ELISA are commercially available, but no con-
samples, since, especially in direct, non-extraction
vincing evidence of their claimed specificity is pro-
methods, the matrix in which the calibrators are
vided by the manufacturer. The use of such methods
dissolved may be inappropriate, resulting in non-
cannot be recommended.
linearity. The serummatrix effect can be avoided
using extraction methods.
4.5 Quality control Commercial kits contain control sera. Kit controls
Overall, commercially available kits for testosterone usually perform quite well and provide results in the
measurement perform fairly well in the male, eugo- expected range. However, care should be taken to use
nadal range; while inaccuracy and imprecision are a independent, certified control sera, in which the tes-
major problem at low concentrations. This prompted tosterone concentration has been determined by MS.
a group of stakeholders to convene a meeting to reach Quite often kit controls are in the expected range,
consensus about the urgent need for improvement in while certified control sera are not. Such kits should
testosterone testing (Rosner et al. 2010). In essence, not be used. Another criterion for choice is compari-
the recommendations acknowledged the lack of son of results in a sufficient number of patient sera to
standardization of actual testosterone assays, pleading those obtained with a validated method. All these tests
for definition of acceptable performance criteria, should be performed even if the candidate testoster-
appropriate reference intervals and guidelines for one system is part of an automatic multianalyzer.
patient preparation and sample handling. In addition, Companies should be asked to contribute data on
healthcare providers, research organizations and sci- the calibrators and to allow complete validation
entific journals are to be stimulated to promote, reim- before the choice to adopt that system is made. This
burse and accept only testosterone determinations is particularly relevant considering the overall poor
performed with standardized methods; the latter to performance in external quality control trials of most
be developed with the active contribution of the kits and automatic analyzers with sera containing low
manufacturers. Given the limitations of the current testosterone concentrations i.e. from children,
commercially available kits, the future seems to be women and hypogonadal men.
irreversibly oriented towards adoption of MS-based Laboratories must often face the decision of
methods, which, however, are still unaffordable for whether to adopt a new methodology. This is due to
many small laboratories for both technical and the large number of kits competing for the market or
budgetary reasons. In the meantime each laboratory to the launch of an improved kit version, to budget-
should carefully revise the performance of its assay. ary necessities calling for automation and/or adoption
Some suggestions are given hereunder. of a multianalyzer, or to more stringent regulatory

79
Chapter 4: Measuring testosterone, DHT and SHBG

29.70 Fig. 4.1 Internal quality control. Results


Lot 40110 obtained measuring a certified control
serum over one year by enzyme
immunoassay (EIA). Shortly after
23.80 introduction of this control serum it
Testosterone (nmol/l)

became evident that the assay had the


tendency to overestimate the target
value of this lot (17.9 nmol/l). After
servicing of the pipettor/plate reader
17.90
equipment and re-validation (August
2002), the results improved.

12.00

6.10
02

02

02

02

02

02

02

03

03

03

03

03
21

21

21

21

21

21

21

21

21

21
l2

r2
n

ct

ar

ay
Ju

Ap
No

De
Ju

Au

Se

Ja

Fe
O

M
Date

and environmental laws. All these aspects must be duplicate, since the intraduplicate variability at low
considered, but converting to a new methodology concentrations is often >10%.
should never be at the cost of accuracy and reproduci- Another important parameter is assay drift. By
bility. Sometimes changes in reagent lots cause major measuring control sera at the beginning and at the
differences in the results obtained with a kit; these end of the assay, it can be determined whether the
become evident both from the clinical plausibility of results at the end of the assay have any tendency to be
the data and from internal quality control. In this case systematically over- or underestimated. This is par-
a full re-validation should be performed, the kit ticularly relevant in ELISAs and methods relying on
manufacturer approached and, if the problem per- colorimetric reactions. If such a tendency is dis-
sists, the methodology changed. covered, the number of samples which can be meas-
ured in one assay should be reduced. As a general
rule, if the assay embraces several microtiter plates,
4.5.2 Internal quality control then calibrators or even entire standard curves should
Once a system has been adopted, some long-term be run in each plate.
parameters have to be kept under constant control. Inter-assay precision is checked by using certi-
These parameters include intra- and inter-assay vari- fied control sera with three different testosterone
ability, assay drift, maximal and non-specific binding, concentrations (low, middle, high) in each assay.
and the standard curve characteristics, such as the Changes in control sera lots have to be recorded.
slope and the dose at 50% curve displacement. Con- The results obtained in each assay must fall within
trol charts have to be set up and constantly observed a predetermined, allowed range, usually within 2
in order to distinguish between sporadic and system- standard deviations (SDs) of the target value, other-
atic changes in some parameters; the latter being an wise repeating the assay should be considered.
indication of some modification in the kit (e.g. a Control sera can give results outside the allowed
change in a reagent lot) or some problem in the range sporadically, but ideally their variability
equipment. Intra-assay precision is evaluated by cal- should oscillate equally on the two sides of the
culating the variability of the duplicate determin- target value. The persistence of results mainly on
ations. Measuring samples in singlicate does not one side indicates a systematic problem, which, in
allow evaluation of this parameter. In our experience the example of Fig. 4.1 was identified in the auto-
testosterone and DHT should always be measured in matic pipettor.

80
Chapter 4: Measuring testosterone, DHT and SHBG

Fig. 4.2 External quality assessment.


RIA EIA Results of two control sera with a
testosterone concentration of 12.2 nmol/l
20.3 20.3 (sample A, y-axis) and 40.9 nmol/l (sample
17.1 17.1 B, x-axis), respectively as measured by MS
in German laboratories participating in an
12.2 EQA scheme. Results are grouped by
12.2 radioimmunoassay (RIA), and methods
based on enzymatic (EIA), fluorimetric
7.32 (FIA) or luminescent (LIA) detection
4.07 4.07 7.32 systems. Each dot represents results from
one laboratory. Target values are
24.5 57.3 24.5 57.3 indicated by the small white square at the
center of each graph. According to
13.5 40.9 68.2 13.5 40.9 68.2 current German guidelines the target is
considered to be successfully met if the
FIA LIA results fall within 40% of the value
measured by MS, indicated by the square
20.3 20.3 defined by dotted lines. Results available
17.1 17.1 online from the German Society of
Clinical Chemistry (www.dgkc-online.de).
12.2 12.2

4.07 7.32 4.07 7.32

24.5 57.3 24.5 57.3


13.5 40.9 68.2 13.5 40.9 68.2

practice this means that a man with borderline serum


4.5.3 External quality assessment testosterone concentrations has an equal probability
In most western countries, successful participation in of being classified as normal or hypogonadal (i.e. > or
an external quality assessment (EQA) program is a < 12 nmol/l), and both diagnoses are correct from the
prerequisite to obtain and maintain the license to analytical point of view. The comparison between the
perform diagnostic tests for many analytes, including four method groups shown in Fig. 4.2 does not reveal
testosterone. Participating laboratories receive major differences among them.
samples with unknown testosterone concentrations In other countries, as in the USA, proficiency of
and return the results to the scheme organizer. The testosterone assessment is not evaluated by compar-
results are then evaluated and the laboratory receives ing the results of the control sera distributed to those
an assessment of its performance. An example of the obtained by MS-based methods but just by assessing
results of the EQA survey in Germany, organized by the consensus between all laboratories using the same
the German Society of Clinical Chemistry is given in methodology/instrument. An illustrative example of
Fig. 4.2, showing the very high inter-laboratory vari- this is provided by Rosner et al. (2007), and highlights
ability of the results obtained from the measurement the consequences of this lack of standardization.
of two serum samples containing testosterone con- Therefore, as long as commercially available kits are
centrations in the adult male range. Most of the used, the accuracy of the serum testosterone deter-
laboratories manage to produce results falling within mination is very much dependent on the in-house re-
the allowed range, which, according to the actual validation and on very strict internal quality control;
German guidelines, permits variations of 40% of both of which remain up to the individual laboratory.
the testosterone value measured by MS. This is, of External quality assessment for SHBG and free
course, a very wide range, such that the measurement testosterone is much less advanced. The UK NEQAS
of sample A of Fig. 4.2, which has a nominal value of (National External Quality Assessment Service) and
12.2 nmol/l, is considered successful if the laboratory US CAP (College of American Pathologists) offer
obtains any value between 7.32 and 17.1 nmol/l. In schemes for SHBG, but only a few laboratories make

81
Chapter 4: Measuring testosterone, DHT and SHBG

use of them. The US CAP survey 2003 reports on 79


laboratories measuring SHBG and 9 laboratories  Non-extraction, direct methods based on
non-radioactively labeled tracers are currently
measuring bioavailable testosterone by ammonium
routinely used. Automatic multianalyzers are the
sulfate precipitation. As far as free testosterone is
systems mostly used for serum testosterone
concerned, 6 laboratories were reported to use equi- measurement. These methods show
librium dialysis, 3 laboratories centrifugal ultrafiltra- unacceptably high inaccuracy.
tion and 70 laboratories used an analog method. In  Any testosterone measurement system,
view of the analytical problem of the analog kits including those based on automatic analyzers,
reported above, it is not surprising that these labora- should be carefully validated in house against an
tories produce free testosterone results much too low extraction method before it is adopted for
compared to those obtained by free testosterone cal- routine assays.
culation, dialysis or ultrafiltration. No external quality  Carefully validated immunological methods
assessment is presently available for DHT. perform reasonably well in the male, eugonadal
range.
4.6 Key messages  In general, all methods currently in use suffer
from poor standardization.
 Serum testosterone is measured in clinical So-called analog free testosterone methods are

routine by immunological competitive methods
unreliable. Calculated free testosterone is very
based on polyclonal antisera and labeled
popular, but the best estimation of free
hormone.
testosterone is given by dialysis or ultrafiltration.
 The reference method for testosterone
 DHT should be measured by chromatographic or
measurement is MS. A number of LC-MS/MS
oxidative methods. Mass spectrometric methods
methods have been validated for accurate
are under development.
assessment of testosterone. Extraction methods
 Participation in external quality control programs
should be used for accurate, reproducible
is mandatory. Strict internal quality control is
testosterone measurements whenever MS-based
fundamental to ensure accurate measurements.
methods are not possible.

4.7 References Column-switching LC-MS/MS


analysis for quantitative
Ceglarek U, Kortz L, Leichtle A, Fiedler
GM, Kratzsch J, Thiery J (2009)
Anderson DC (1974) Sex-hormone- determination of testosterone in Rapid quantification of steroid
binding globulin. Clin Endocrinol human serum. Clin Chim Acta patterns in human serum by on-line
(Oxf) 3:6996 382:134137 solid-phase extraction combined
Atkinson KR, Lo KR, Payne SR, with liquid chromatography-triple
Brandhorst G, Streit F, Kratzsch J,
Mitchell JS, Ingram JR (2008) Rapid quadrupole linear ion trap mass
Schiettecatte J, Roth HJ, Luppa PB,
saliva processing techniques for spectrometry. Clin Chim Acta
Krner A, Kiess W, Binder L,
near real-time analysis of salivary
Oellerich M, von Ahsen N 401:114118
steroids and protein. J Clin Lab Anal Chen Y, Yazdanpanah M, Hoffman
(2011) Multicenter evaluation
22:395402 BR, Diamandis EP, Wong PY
of a new automated
Booth A, Johnson DR, Granger DA, electrochemiluminescence (2009) Rapid determination of
Crouter AC, McHale S (2003) immunoassay for the quantification serum testosterone by liquid
Testosterone and child and of testosterone compared to liquid chromatography-isotope dilution
adolescent adjustment: the chromatography tandem mass tandem mass spectrometry and a
moderating role of parentchild spectrometry. Clin Biochem split sample comparison with three
relationships. Dev Psychol 44:264247 automated immunoassays. Clin
39:8598 Biochem 42:484490
Cawood ML, Field HP, Ford CG,
Boots LR, Potter S, Potter D, Azziz R Gillingwater S, Kicman A, Cowan D, Chen Y, Yazdanpanah M, Wang XY,
(1998) Measurement of total serum Barth JH (2005) Testosterone Hoffman BR, Diamandis EP, Wong
testosterone levels using measurement by isotope-dilution PY (2010) Direct measurement of
commercially available kits: high liquid chromatography-tandem serum free testosterone by
degree of between-kit variability. mass spectrometry: validation ultrafiltration followed by liquid
Fertil Steril 69:286292 of a method for routine chromatography tandem mass
Borrey D, Moerman E, Cockx A, clinical practice. Clin Chem spectrometry. Clin Biochem
Engelrelst V, Langlois MR (2007) 51:14721479 43:490496

82
Chapter 4: Measuring testosterone, DHT and SHBG

Chiappin S, Antonelli G, Gatti R, De Fitzgerald RL, Herold DA (1996) derivatization. Clin Chim Acta
Palo EF (2007) Saliva specimen: a Serum total testosterone: 409:7884
new laboratory tool for diagnostic immunoassay compared with
Janzen N, Sander S, Terhardt M,
and basic investigation. Clin Chem negative chemical ionization gas
Peter M, Sander J (2008) Fast and
Acta 383:3040 chromatography-mass
direct quantification of adrenal
Dabbs JM Jr (1990) Salivary spectrometry. Clin Chem
steroids by tandem mass
testosterone measurements: 42:749755
spectrometry in serum and dried
reliability across hours, days, and Fitzgerald RL, Griffin TL, Herold DA blood spots. J Chromatogr B Analyt
weeks. Physiol Behav 48:8386 (2010) Analysis of testosterone in Technol Biomed Life Sci 861:117122
Davies R, Collier C, Raymond M, serum using mass spectrometry.
Jockenhvel F, Krsemann C, Jaeger A,
Heaton J, Clark A (2002) Indirect Methods Mol Biol 603:489500
Olbricht T, Reinwein D (1992)
measurement of bioavailable Fritz KS, McKean AJ, Nelson JC, Comparability of serum
testosterone with the Bayer Immuno Wilcox RB (2008) Analog-based testosterone determination with the
1 system. Clin Chem 48:388490 free testosterone test results linked ten most commonly used
Dechaud H, Lejeune H, Garoscio- to total testosterone concentrations, commercial radioimmunoassays
Cholet M, Mallein R, Pugeat M not free testosterone concentrations. and with a new enzyme
(1989) Radioimmunoassay of Clin Chem 54:512516 immunoassay. Klin Lab 38:8188
testosterone not bound to sex- Furuyama S, Mayes DM, Nugent CA Kalhorn TF, Page ST, Howald WN,
steroid-binding protein in plasma. (1970) A radioimmunoassay for Mostaghel EA, Nelson PS (2007)
Clin Chem 35:16091614 plasma testosterone. Steroids Analysis of testosterone and
De Ronde W, van der Schouw YT, Pols 16:415428 dihydrotestosterone from biological
HA, Gooren LJ, Muller M, Grobbee Gonzalez-Sagrado M, Martin-Gil FJ, fluids as the oxime derivatives using
DE, de Jong FH (2006) Calculation of Lopez-Hernandez S, Fernandez- high-performance liquid
bioavailable and free testosterone in Garcia N, Olmos-Linares A, chromatography/tandem mass
men: a comparison of 5 published Arranz-Pena ML (2000) Reference spectrometry. Rapid Commun Mass
algorithms. Clin Chem 52:17771784 values and methods comparison of Spectrom 21:32003206
Dunn JF, Nisula BC, Rodbard D (1981) a new testosterone assay on the Kane J, Middle J, Cawood M (2007)
Transport of steroid hormones: AxSYM system. Clin Biochem Measurement of serum testosterone
binding of 21 endogenous steroids 33:175179 in women; what should we do? Ann
to both testosterone-binding Granger DA, Shirtcliff EA, Booth A, Clin Biochem 44:515
globulin and corticosteroid-binding Kivlighan KT, Schwartz EB
Krone N, Hughes BA, Lavery GG,
globulin in human plasma. J Clin (2004) The trouble with
Stewart PM, Arlt W, Shackleton CH
Endocrinol Metab 53:5868 salivary testosterone.
(2010) Gas chromatography/mass
Dussault JH, Letarte J, Guyda H, Psychoneuroendocrinology
spectrometry (GC/MS) remains a
Laberge C (1977) Serum thyroid 29:12291240
pre-eminent discovery tool in
hormone and TSH concentrations Guo T, Chan M, Soldin SJ (2004) clinical steroid investigations even
in newborn infants with congenital Steroid profiles using liquid in the era of fast liquid
absence of thyroxine-binding chromatography-tandem mass chromatography tandem mass
globulin. J Pediatr 90:264265 spectrometry with atmospheric spectrometry (LC/MS/MS). J Steroid
Ekins R (1990) Measurement of free pressure photoionization source. Biochem Mol Biol 121:496504
hormones in blood. Endocr Rev Arch Pathol Lab Med 128:469475
Kulle AE, Riepe FG, Melchior D, Hiort
11:546 Guo T, Taylor RL, Singh RJ, Soldin SJ O, Holterhus PM (2010) A novel
Fanelli F, Belluomo I, Di Lallo VD, (2006) Simultaneous determination ultrapressure liquid
Cuomo G, De Iasio R, Baccini M, of 12 steroids by isotope dilution chromatography tandem mass
Casadio E, Casetta B, Vicennati V, liquid chromatography-photospray spectrometry method for the
Gambineri A, Grossi G, Pasquali R, ionization tandem mass simultaneous determination of
Pagotto U (2011) Serum steroid spectrometry. Clin Chim Acta androstenedione, testosterone, and
profiling by isotopic dilution- 372:7682 dihydrotestosterone in pediatric
liquid chromatography-mass Harwood DT, Handelsman DJ (2009) blood samples: age- and sex-specific
spectrometry: comparison Development and validation of a reference data. J Clin Endocrinol
with current immunoassays sensitive liquid chromatography- Metab 95:23992409
and reference intervals in tandem mass spectrometry assay to Kushnir MM, Rockwood AL, Roberts
healthy adults. Steroids simultaneously measure androgens WL, Pattison EG, Bunker AM,
76:244253 and estrogens in serum without Fitzgerald RL, Meikle AW (2006)

83
Chapter 4: Measuring testosterone, DHT and SHBG

Performance characteristics of a serum to support testosterone Moal V, Mathieu E, Reynier P,


novel tandem mass spectrometry replacement therapy for Malthiry Y, Gallois Y (2007) Low
assay for serum testosterone. Clin hypogonadism. Steroids 73:601610 serum testosterone assayed by liquid
Chem 52:120128 Ly LP, Sartorius G, Hull L, Leung A, chromatography-tandem mass
Kushnir MM, Blamires T, Rockwood Swerdloff RS, Wang C, Handelsman spectrometry. Comparison with five
AL, Roberts WL, Yue B, Erdogan E, DJ (2010) Accuracy of calculated immunoassay techniques. Clin
Bunker AM, Meikle AW (2010) free testosterone formulae in Chim Acta 386:1219
Liquid chromatography- men. Clin Endocrinol (Oxf) Morley JE, Perry HM 3rd, Patrick P,
tandem mass spectrometry 73:382388 Dollbaum CM, Kells JM (2006)
assay for androstenedione, Macdonald PR, Owen LJ, Wu FC, Validation of salivary testosterone
dehydroepiandrosterone, and Macdowall W, Keevil BG; NATSAL as a screening test for male
testosterone with pediatric and Team (2011) A liquid hypogonadism. Aging Male
adult reference intervals. Clin Chem chromatography-tandem mass 9:165169
56:11381147 spectrometry method for salivary Need EF,OLoughlin PD, Armstrong
Kushnir MM, Rockwood AL, Roberts testosterone with adult male DT, Haren MT, Martin SA, Tilley
WL, Yue B, Bergquist J, Meikle AW reference interval determination. WD; the Florey Adelaide Male
(2011) Liquid chromatography Clin Chem 57:774775 Aging Study, Witter GA, Buchanan
tandem mass spectrometry for Manni A, Pardridge WM, Cefalu W, G (2010) Serum testosterone
analysis of steroids in clinical Nisula BC, Bardin CW, Santner SJ, bioassay evaluation in a large male
laboratories. Clin Biochem 44:7788 Santen RJ (1985) Bioavailability of cohort. Clin Endocrinol 72:8798
Legro RS, Schlaff WD, Diamond MP, albumin-bound testosterone. J Clin Nieschlag E, Loriaux DL (1972)
Coutifaris C, Casson PR, Brzyski Endocrinol Metab 61:705710 Radioimmunoassay for plasma
RG, Christman GM, Trussell JC, Matsui F, Koh E, Yamamoto K, testosterone. Z Klin Chem Klin
Krawetz SA, Snyder PJ, Ohl D, Sugimoto K, Sin HS, Maeda Y, Biochem 10:164168
Carson SA, Steinkampf MP, Carr Honma S, Namiki M (2009) Liquid Rassaie MJ, Kumari LG, Pandey PK,
BR, McGovern PG, Cataldo NA, chromatography-tandem mass Gupta N, Kochupillai N, Grover PK
Gosman GG, Nestler JE, Myers ER, spectrometry (LC-MS/MS) assay for (1992) A highly specific
Santoro N, Eisenberg E, Zhang M, simultaneous measurement of heterologous enzyme-linked
Zhang H; Reproductive Medicine salivary testosterone and cortisol in immunosorbent assay for
Network (2010) Total testosterone healthy men for utilization in the measuring testosterone in plasma
assay in women with polycystic diagnosis of late-onset using antibody-coated
ovary syndrome: precision and hypogonadism in males. Endocr J immunoassay plates or
correlation with hirsutism. J Clin 56:10831093 polypropylene tubes. Steroids
Endocrinol Metab 95:53055313 57:288294
Mendel CM (1992) The free hormone
Levesque A, Letellier M, Swirski C, hypothesis. Distinction from the Raverot V, Lopez J, Grenot C, Pugeat
Lee C, Grant A (1998) Analytical free hormone transport hypothesis. M, Dchaud H (2010) New
evaluation of the testosterone assay J Androl 13:107116 approach for measurement of non-
on the Bayer Immuno 1 system. SHBG-bound testosterone in
Mendel CM, Murai JT, Siiteri PK,
Clin Biochem 31:2328 human plasma. Anal Chim Acta
Monroe SE, Inoue M (1989)
Levine A, Zagoory-Sharon O, Conservation of free but not total or 658:8790
Feldman R, Lewis JG, Weller A non-sex-hormone-binding- Rauh M (2010) Steroid measurement
(2007) Measuring cortisol in human globulin-bound testosterone in with LC-MS/MS. Application
psycobiological studies. Physiol serum from Nagase analbuminemic examples in pediatrics. J Steroid
Behav 30:4353 rats. Endocrinology 124:31283130 Biochem Mol Biol 121:520527
Lewis JG (2006) Steroid analysis in Middle JG (2002) UK NEQAS for Rauh M, Grschl M, Rascher W, Drr
saliva: an overview. Clin Biochem Steroid Hormones Annual Review HG (2006) Automated, fast and
Rev 27:139146 2002. UK NEQAS, University sensitive quantification of 17 alpha-
Licea-Perez H, Wang S, Szapacs ME, Hospital Birmingham NHS Trust hydroxy-progesterone,
Yang E (2008) Development of a Middle JG (2007) androstenedione and testosterone
highly sensitive and selective UPLC/ Dehydroepiandrosterone by tandem mass spectrometry with
MS/MS method for the sulphate interferes in many on-line extraction. Steroids
simultaneous determination of direct immunoassays for 71:450458
testosterone and 5alpha- testosterone. Ann Clin Biochem Rosner W (1997) Errors in the
dihydrotestosterone in human 44:173177 measurement of plasma free

84
Chapter 4: Measuring testosterone, DHT and SHBG

testosterone. J Clin Endocrinol Shirtcliff EA, Granger DA, Schwartz E, Best Pract Res Clin Endocrinol
Metab 82:20142015 Curran MJ (2001) Use of salivary Metab 20:1533
Rosner W (2001) An extraordinarily biomarkers in biobehavioral Swerdloff RS, Wang C (2008) Free
inaccurate assay for free research: cotton-based sample testosterone measurement by the
testosterone is still with us. J Clin collection methods can interfere analog displacement direct assay:
Endocrinol Metab 86:2903 with salivary immunoassay results. old concerns and new evidence. Clin
Psychoneuroendocrinology Chem 54:458460
Rosner W, Vesper H on behalf of the 26:165173
Endocrine Society and the Tai SS, Xu B, Welch MJ, Phinney KW
endorsing organizations (2010) Shirtcliff EA, Granger DA, Likos A (2007) Development and evaluation
Toward excellence in testosterone (2002) Gender differences in the of a candidate reference
testing: a consensus statement. validity of testosterone measured in measurement procedure for the
J Clin Endocrinol Metab saliva by immunoassay. Horm determination of testosterone in
95:45424548 Behav 42:6269 human serum using isotope dilution
Rosner W, Hryb DJ, Khan MS, Nakhla Simoni M (2004) Methodology for liquid chromatography/tandem
AM, Romas NA (1999) Sex measuring testosterone, DHT and mass spectrometry. Anal Bioanal
hormone-binding globulin mediates SHBG in a clinical setting. In: Chem 388:10871094
steroid hormone signal Nieschlag E, Behre HM (eds) Taieb J, Benattar C, Birr AS,
transduction at the plasma Testosterone: Action, Deficiency, Lindenbaum A (2002) Limitations
membrane. J Steroid Biochem Mol Substitution, 3rd edn. Cambridge of steroid determination by direct
Biol 69:481485 University Press, Cambridge, immunoassay. Clin Chem
pp 641664 48:583585
Rosner W, Auchus RJ, Azziz R, Sluss
PM, Raff H (2007) Position Simoni M, Nieschlag E (2010) Taieb J, Mathian B, Millot F, Patricot
statement: utility, limitations and Endocrine laboratory diagnosis. In: MC, Mathieu E, Queyrel N, Lacroix
pitfalls in measuring testosterone: Nieschlag E, Behre HM, Nieschlag S I, Somma-Delpero C, Bouloux P
an Endocrine Society position (eds) Andrology. Male (2003) Testosterone measured by 10
statement. J Clin Endocrinol Metab Reproductive Health and immunoassays and by isotope-
92:405413 Dysfunction, 3rd edn. Springer- dilution gas chromatography-mass
Verlag, Berlin, pp 109118 spectrometry in sera from 116 men,
Roy P, Alevizaki M, Huhtaniemi I
(2008) In vitro bioassays for Simoni M, Baraldi E, Baraghini GF, women and children. Clin Chem
androgens and their diagnostic Boraldi V, Roli L, Seghedoni S, 49:13811395
applications. Hum Reprod Update Verlardo A, Montanini V (1988) Thienpont LM, Van Uytfanghe K,
14:7382 Twenty-four-hour pattern of Blincko S, Ramsay CS, Xie H, Doss
plasma SHBG, total proteins and RC, Keenl BG, Owen LS, Rockwood
Salameh WA, Redor-Goldman MM,
testosterone in young and elderly AL, Kushnir MM, Chun KY,
Clarke NJ, Reitz RE, Caulfield MP
men. Steroids 52:381382 Chandler DW, Field HP, Sluss PM
(2010) Validation of a total
testosterone assay using high- Simoni M, Montanini V, Fustini MF, (2008) State-of-the-art of serum
turbulence liquid chromatography Del Rio G, Cioni K, Marrama P testosterone measurement by
tandem mass spectrometry: total (1992) Circadian rhythm of plasma isotope dilution-liquid
and free testosterone reference testosterone in men with idiopathic chromatography-tandem mass
ranges. Steroids 75:169175 hypogonadotrophic hypogonadism spectrometry. Clin Chem
before and during pulsatile 54:12901297
Shackleton C (2010) Clinical steroid
administration of gonadotropin- Turpeinen U, Linko S, Itkonen O,
mass spectrometry: a 45-year
releasing hormone. Clin Endocrinol Hmlinen E (2008)
history culminating in HPLC-MS/
36:2934 Determination of testosterone in
MS becoming an essential tool for
patient diagnosis. J Steroid Biochem Singh RJ (2008) Validation of a high serum by liquid chromatography-
Mol Biol 121:481490 throughput method for serum/ tandem mass spectrometry. Scand
plasma testosterone using liquid J Clin Lab Invest 68:5057
Shiraishi S, Lee PW, Leung A, Goh VH,
Swerdloff RS, Wang C (2008) chromatography tandem mass Vaitukaitis J, Robbins JB, Nieschlag E,
spectrometry (LC-MS/MS). Steroids Ross GT (1971) A method for
Simultaneous measurement of
serum testosterone and 73:13391344 producing specific antisera with
dihydrotestosterone by liquid Soto AM, Maffini MV, Schaeberle CM, small doses of immunogen. J Clin
chromatography-tandem mass Sonnenschein C (2006) Strengths Endocrinol Metab 33:988991
spectrometry. Clin Chem and weaknesses of in vitro assays for Van Uytfanghe K, Stckl D, Kaufman
54:18551863 estrogenic and androgenic activity. JM, Fiers T, Ross HA, De

85
Chapter 4: Measuring testosterone, DHT and SHBG

Leenheer AP, Thienpont LM (2004) (1982) An improved ultrafiltration testosterone. Ann Clin Biochem
Evaluation of a candidate reference method for determining free 43:196199
measurement procedure for serum testosterone in serum. Clin Chem Winters SJ, Kelley DE, Goodpaster B
free testosterone based on 28:22862291 (1998) The analog free
ultrafiltration and isotope dilution- Wang C, Plymate S, Nieschlag E, testosterone assay: are the results in
gas chromatography-mass Paulsen CA (1981) Salivary men clinically useful? Clin Chem
spectrometry. Clin Chem testosterone in men: further 44:21782182
50:21012110 evidence of a direct correlation Wolthers BG, Kraan GP (1999)
Vermeulen A, Verdonck L, Kaufman with free serum testosterone. Clinical applications of gas
JM (1999) A critical evaluation of J Clin Endocrinol Metab chromatography and gas
simple methods for the estimation 53:10211024 chromatography-mass spectrometry
of free testosterone in serum. J Clin Wang C, Catlin DH, Demers LM, of steroids. J Chromatogr A
Endocrinol Metab 84:36663672 Starcevic B, Swerdloff RS (2004) 843:247274
Vesper HW, Bhasin S, Wang C, Tai SS, Measurement of total serum Yamashita K, Miyashiro Y, Maekubo
Dodge LA, Singh RJ, Nelson G, testosterone in adult men: H, Okuyama M, Honma S,
Ohorodnik S, Clarke NJ, Salameh comparison of current laboratory Takahashi M, Numazawa M (2009)
WA, Parker CR Jr, Razdan R, methods versus liquid Development of highly
Mansell EA, Myers GL (2009) chromatography-tandem mass sensitive quantification method
Interlaboratory comparison study spectrometry. J Clin Endocrinol for testosterone and
of serum total testosterone Metab 89:534543 dihydrotestosterone in human
[corrected] measurements Warner MH, Kane J, Atkin SL, serum and prostate tissue by
performed by mass spectrometry Kilpatrick ES (2006) liquid chromatography-
methods. Steroids 74:498503 Dehydroepiandrosterone sulphate electrospray ionization tandem
Vlahos I, MacMahon W, Sgoutas D, interferes with the Abbott Architect mass spectrometry. Steroids
Bowers W, Thompson J, Trawick W direct immunoassay for 74:920926

86
Chapter
The behavioral correlates of testosterone

5 John Bancroft

5.1 Introduction 87 Antiandrogens in women 101


5.2 Testosterone and gender differentiation 88 Bilateral ovariectomy 102
5.2.1 Gender differentiation of the brain 88 5.4.1.4 The effects of exogenous androgen
administration on the sexuality of
5.2.2 The mode of action of androgens and
women 103
estrogens in the brain 89
Testosterone administration to
5.3 Activational effects of testosterone healthy premenopausal women 103
and behavior in adult men 90 Premenopausal women with sexual
5.3.1 Sexual desire and response 90 problems 103
5.3.1.1 Testosterone replacement in Premenopausal women with
hypogonadal men 90 endocrine abnormalities 104
5.3.1.2 Experimental reduction of Naturally postmenopausal
testosterone in eugonadal men 92 women 104
5.3.2 Testosterone, mood and aggression Surgical menopause 105
in men 93 5.4.2 Testosterone, mood and aggression
5.3.2.1 Depression 93 in women 106
5.3.2.2 Aggression 93 5.4.2.1 Mood 106
5.3.2.3 Testosterone and sleep 96 5.4.2.2 Aggression and assertiveness 108
5.3.3 Summary of evidence in the male 97 5.4.3 Summary of evidence in the female 109
5.4 Activational effects of testosterone and 5.5 Comparison of testosterone effects in men
behavior in adult women 98 and women 110
5.4.1 Sexual desire and response 98 5.5.1 Some hypothetical ideas about
5.4.1.1 Circulating androgen levels and their testosterone and gender
relation to the sexuality of differences 112
women 98 5.5.1.1 The Desensitization hypothesis 113
The menstrual cycle 99 5.5.1.2 The By-product hypothesis 113
5.4.1.2 Aging and the menopause 100 5.5.1.3 One way forward 114
5.4.1.3 Effects of iatrogenic lowering of
5.6 Key messages 114
testosterone in women 100
5.7 References 115
Oral contraceptives 100

5.1 Introduction most aspects of the human condition, scientific


When the two principal sex hormones, testosterone research has continued to uncover the complexity of
and estradiol, were first identified in the 1940s, they sex hormone function, to the extent that our ability to
provided us with a simple distinction between a male comprehend is being challenged. In this chapter we
hormone and a female hormone. Since then, as with will be focusing on the relationships between

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

87
Chapter 5: The behavioral correlates of testosterone

testosterone and various aspects of our behavior; hormones. Early exposure of the brain to hormones
hence we will be principally concerned with hor- results in organization of its structure and responsive-
mone-related brain mechanisms that underlie those ness to the activational effects of the same hormones
behaviors. We now know that testosterone and estra- later in development. In lower animals, particularly
diol have an important role in both male and female rodents, there are clear differences between male and
brains, and that testosterone is converted to estradiol, female brains determined by the organizational effects
leaving us unsure of how many correlates of testoster- of both testosterone and estradiol. These effects are
one are in fact primarily correlates of estradiol. There probably mediated mainly by hormonal influence on
is now evidence to suggest that, much earlier in our apoptosis, or programmed cell death, during brain
phylogenetic history, testosterone was a precursor of development (Gorski 2000).
estradiol (Thornton 2001). We have to grapple with In the human, the picture is less clear and has
the fact that if testosterone influences the behavior of resulted in much controversy (reviewed by Hines
women, it does so with levels of testosterone in the 2004). Gender differences in the size and shape of
blood around a 10th of those found in men. We also the corpus callosum, the main connecting pathway
have a variety of androgenic hormones in addition between the right and left hemispheres, have been
to testosterone to consider. In particular, DHEA and found in some studies, but not all. In any case, such
its sulfate, DHEAS, which are mainly produced by differences are probably most relevant to certain
the adrenal glands, are present in the blood at levels aspects of cognitive function, and not to gender-related
substantially higher than testosterone or other andro- or sexual behavior. The anterior hypothalamic/
gens, yet their function is not well understood. There preoptic area, which contains many testosterone and
is relatively recent evidence of synthesis of androgenic estradiol receptors, is of interest. This area has four
hormones within brain cells, these being examples of sub-regions, called the interstitial nuclei of the anter-
what are now known as neurosteroids, not derived ior hypothalamus (INAH 14). Evidence of gender
from the classical hormones in the blood (e.g. King difference has only been found in INAH-3. This is
2008). The original assumption that testosterone has consistently larger and contains more neurons in men
direct effects only at genomic receptors is being chal- than in women; though the behavioral significance of
lenged, and the genetic variability of ARs is now very this is not yet clear. The central region of the bed
evident. We will encounter most of these complexities nucleus of the stria terminalis (BNST), which also
as we progress through this chapter. contains steroid-concentrating neurons (Hines 2004),
The evidence will be reviewed under three principal is smaller in women than in men. This was also found to
headings: (1) testosterone and gender differentiation; (2) be smaller in six male-to-female transsexuals, leaving us
activational effects of testosterone in men; and (3) acti- uncertain as to whether this indicates a structural
vational effects of testosterone in women. Whereas the difference that may have contributed to their transgen-
principal behaviors and associated physiologic responses der identity, or whether it was a consequence of the
to be considered relate to sex, the relevance of testoster- administration of estradiol that is used by male-to-
one to mood and aggression, and how such relationships female transsexuals to produce feminizing bodily effects
may interact with sex, will also be discussed. (Zhou et al. 1995). This raises the question of whether
organizing effects of testosterone and estradiol on the
brain are restricted to specific stages of human brain
5.2 Testosterone and gender development, as they clearly are in rodents. These
differentiation specific brain areas showing gender difference are too
small to be examined functionally with brain imaging,
5.2.1 Gender differentiation of the brain and the evidence of their existence is so far based on
Whereas the role of testosterone, and its 5-a-reduced post-mortem examination.
form, DHT, in determining the male anatomical Our understanding of human brain development
development of the fetus, is reasonably clear (see has, however, changed substantially in the past decade
Chapter 2), the impact of testosterone on gender as a result of structural and functional brain imaging
differentiation of brain development is less well studies. It is now clear that there are major changes in
understood. Here we need to distinguish between the brain during the second decade of life (Weinberger
organizational and activational effects of et al. 2005). These are likely to be affected by hormonal

88
Chapter 5: The behavioral correlates of testosterone

changes around puberty, and hence will vary according least one type of ER to show normal sexual function.
to whether puberty is early, late or at the normal time. The relevance of the ERs to the sexuality of female
The timing of human puberty can vary substantially, mice has not yet been clarified. In the human we have
with trends towards earlier puberty having occurred in no directly comparable evidence.
certain parts of the world, and with evidence suggest- The conventional wisdom is that testosterone acts
ing that this reflects an interaction between genetic in the brain via genomic effects (Rommerts 1990);
factors and environmental conditions (Parent et al. whereas estradiol and progesterone, in addition to
2003). It is also possible that adrenarche, which is such intracellular genomic effects, can also have much
confined to humans and primates, may also be more rapid effects by acting on the cell membrane
involved in this longer-term brain organization by estradiol being excitatory and progesterone inhibitory.
androgens, although this has not yet been studied. However, recent animal research has shown evidence
Perrin et al. (2008), using magnetic resonance of ARs in axons, dendrites and glial processes in the
imaging (MRI) scanning in a cross-sectional study neocortex, hippocampus and amygdala. These may be
of adolescent boys and girls, found a clear sexual involved in more rapid behavioral effects of androgens
dimorphism in the increase of brain white matter, (Sarkey et al. 2008). Once again, we have no direct
which was slight in girls and much more marked in evidence of the relevance of this to humans.
boys. They also showed a relationship of this effect Research on the site of action of testosterone in the
to functional polymorphism of the AR gene. It has non-human primate brain goes back more than thirty
previously been shown that a low number of CAG years, and we can assume more direct relevance to the
repeats in this gene (i.e. a short gene) is associated human brain than with rodent research. Michael et al.
with a higher androgenic effect than AR genes with a (1989) used autoradiography to look at both androgen
high number of CAG repeats (a long gene) (e.g. Tut and estrogen activity in male rhesus monkeys. Activity
et al. 1997). Perrin et al. (2008) found the increase in of 5a-reductase, which converts testosterone into
white matter volume was greater in males with the DHT, was fairly evenly distributed in the brain,
short gene. This demonstrates the role of the AR gene whereas aromatase activity, which converts testoster-
in the organizational effects of testosterone. one into estradiol, was much more localized. They
The transcriptional function of the AR gene presents concluded that testosterone acts by being converted
us with a puzzling gender difference. The short gene in to estradiol in the following areas: medial preoptic
the male, as it is more active than the long gene, results in and ventromedial hypothalamic nuclei, the bed
a reduction in plasma testosterone levels; less testoster- nucleus of the stria terminalis (BNST), and the cortical,
one is required to achieve the same effect. In contrast, in medial and accessory amygdaloid nuclei. Testosterone
the female, whereas the short gene is also more active in activity, they concluded, was also mediated by ARs in
its transcriptional function, it is associated with higher the lateral septal, pre-mamillary and intercalated
levels of plasma testosterone, reflecting the gender dif- mamillary nuclei. Roselli et al. (2001), using in situ
ferences in control of androgen production. In the male hybridization histochemistry to locate cytochrome
this is mainly controlled via the hypothalamus. In the P450 aromatase nRNA in the hypothalamus and amyg-
female there are probably direct effects of testosterone dala of cynomolgus monkeys, reported broadly similar
on the adrenal glands and the ovaries (Westberg et al. findings to those of Michael et al. (1989). Abdelgadir
2001). It is not clear what the implications of this are for et al. (1999) used a ribonuclease protection assay to
the organizational effects of testosterone in female brain assess the distribution of AR mRNA in male rhesus
development. These issues about AR gene effects will be monkeys. Again their findings were similar to those of
considered further later in this chapter. Michael et al. (1989). They also compared intact with
castrated males, and found little difference in the distri-
5.2.2 The mode of action of androgens bution of AR mRNA, concluding that, in the brain, AR
is not regulated by androgen at the transcription level.
and estrogens in the brain Two studies of AR distribution in the human male
The current view is that there is one type of AR in the temporal cortex have been reported, both showing sub-
brain, which can be activated by either testosterone or stantial amounts of AR (Sarrieau et al. 1990; Puy et al.
DHT. Recent research with mice has revealed two 1995). In both studies, the temporal lobe was used simply
estrogen receptors, ERa and ERb. Male mice need at because it was available from surgical interventions.

89
Chapter 5: The behavioral correlates of testosterone

We have less evidence for the female brain. Finley An additional limitation is the tendency for
and Kritzer (1999) found no difference between male testosterone levels in the blood to rise after relevant
and female rhesus monkeys in the amount of AR behaviors (e.g. after a man wins at a sporting activity),
protein in the prefrontal cortex. There have been and usually we are left uncertain as to whether this
some post-mortem studies comparing the brains of has any functional relevance. The second and third
men and women. Fernandez-Guasti et al. (2000) com- categories of information allow us to assess the effects
pared the hypothalami of five men and five women. of sustained changes in testosterone availability. The
They found AR immunoreactivity ranging from fourth, the impact of aging in the male, presents a
intense to relatively weak across several areas of the challenge as it faces us with complex interactions
hypothalamus, and in general the level of receptor between different mechanisms and systems that
protein staining was less in the women. This gender are changing as a result of age. This is reviewed in
difference was particularly marked in the lateral and Chapter 16, and also by Zitzmann et al. (2006), and
medial mamillary nuclei; and in the paraventricular will not be considered further in this chapter.
and supraoptic nuclei, staining was only found in the
males. Overall, there was greater individual variability
in the intensity of receptor staining in the women
5.3.1 Sexual desire and response
than in the men. Whereas most of this research has In considering the sexual effects of testosterone, we
focused on the hypothalamus, those studies that have need to identify four components of sexual arousal:
looked at other brain areas, such as the prefrontal information processing, incentive motivation, central
cortex and temporal cortex, have indicated a wider arousal and genital response. The essence of sexual
distribution of ARs in the brain, which raises the desire is incentive motivation linked to processing of
possibility that effects of testosterone in the brain sexually relevant information, with varying degrees of
are not restricted to mechanisms relevant to sexuality central arousal and genital response, which in the
and reproduction, and might include other activa- male is penile erection.
tional effects (Kelly 1991). The normal range of total testosterone in men is
large (1035 nmol/l), and within this range there is
little predictable relationship to measures of sexual
5.3 Activational effects of desire or behavior (Raboch and Starka 1973; Kraemer
et al. 1976). Two studies, in which erectile response to
testosterone and behavior in erotic stimuli was measured, found a correlation
adult men between plasma testosterone and latency but not
There are several types of information of relevance: (1) degree of erectile response (Rubin et al. 1979; Lange
measurement of circulating levels of testosterone, in et al. 1980). It is widely assumed that there is a level of
particular free testosterone, and their relationship to testosterone or threshold, above which increases in
behavioral measures; (2) the consequences of reducing testosterone will make little or no difference to sexual
testosterone levels or blocking testosterone effects, desire or response. This threshold may vary across
either iatrogenically in the course of medical treat- men but will be somewhere within the normal range,
ment, or experimentally; (3) the effects of exogenous and it could increase with age.
testosterone administration, short term (single dose)
and long term; (4) the impact of aging. The first 5.3.1.1 Testosterone replacement
category, relating circulating testosterone levels to in hypogonadal men
behavioral parameters, is the most problematic. The effects of testosterone levels below the normal
As Zitzmann and Nieschlag (2001) noted: When range are demonstrated in men with hypogonadism
interpreting testosterone levels, it is often overlooked who receive TRT (testosterone replacement therapy).
that levels of the hormone in body fluids are only a Most controlled studies have investigated hypogona-
small and transitory step in the cascade of hormone dal men already on testosterone replacement for at
action from production to biological effect. It is least several months, who then have their exogenous
therefore not surprising that correlations between testosterone withdrawn. The effects on sexual para-
plasma testosterone levels and specific behaviors vary meters during a period of this withdrawal are assessed
not only across behaviors but also across studies. and followed by administration of testosterone and

90
Chapter 5: The behavioral correlates of testosterone

placebo, using a double-blind, crossover design from Scotland, the other from Hong Kong (Anderson
(Davidson et al. 1979; Luisi and Franchi 1980; Skak- et al. 1999). There were some interesting differences
kebaek et al. 1981; Salmimies et al. 1982; Kwan et al. between the two groups. Although the effects on
1983; OCarroll et al. 1985). Although most studies of waking erections (i.e. NPT) were similar, the increase
this kind have been small, there has been a reassuring in sexual interest and, more strikingly, masturbation,
consistency in their findings. There is a decline in was less in the Hong Kong men. This confronts us
sexual interest within three to four weeks of androgen with the need to take cultural influences into account.
withdrawal, and if testosterone withdrawal lasts long Nocturnal penile tumescence is unlikely to be cultur-
enough, seminal emission will eventually be impaired. ally sensitive, but masturbation and sexual interest, or
Placebo usually has little effect, but testosterone at least the ways that they are reported and assessed,
replacement restores sexual interest within one or may well be.
two weeks. OCarroll et al. (1985), comparing four Functional brain imaging has also been used to
doses of testosterone undecanoate (40, 80, 120 and compare brain activity in response to sexual stimuli
160 mg daily), showed a clear dose-response relation- in hypogonadal men with and without testosterone
ship in this respect. Effects on sexual activity with replacement. Park et al. (2001), using functional MRI,
ones partner are less consistent, reflecting variable reported on two such men. In both men, activation
partner and relationship characteristics. Masturbation of the inferior frontal lobe, cingulate gyrus, insula
frequency more typically increases. Nocturnal penile and corpus callosum was greater with testosterone
tumescence (NPT) is predictably impaired during replacement. Redout et al. (2005) used positron
testosterone withdrawal and restored with testoster- emission tomography (PET) scanning to compare
one replacement. Nocturnal penile tumescence occurs brain response to sexual stimuli in nine hypogonadal
during REM sleep, which involves a switching off of (H) men, with (H+) and without (H) testosterone
noradrenergic cells in the locus coeruleus in the replacement, and eight eugonadal men. They found
brainstem, which, via their spinal projections, are greater activation in the right orbitofrontal cortex,
responsible for the inhibitory tone in the erectile insula and claustrum in the controls and H+ men
tissues. Thus, in eugonadal men, this reduction in than in the H men. They also found deactivation of
inhibitory tone resulting in erection indicates that the left inferior frontal gyrus, suggestive of reduced
there is normally some degree of excitatory tone. inhibition of sexual arousal, but only in the controls
The locus coeruleus has testosterone receptors (Par- and H+ men. Androgen receptors have been reported
meggiana and Morrison 1990), and this excitatory in the orbitofrontal cortex of primates (Finley and
tone can be considered to be testosterone dependent. Kritzer 1999) and to a limited extent in the cingulate
A few studies of testosterone withdrawal and cortex (Abdelgadir et al. 1999). However, there is,
replacement have used psychophysiologic assessment as yet, no evidence of ARs in the insula or claustrum
of response to visual erotic stimuli. Interestingly, in in primates or humans.
the low testosterone state, erection to such stimuli can A further unresolved issue is whether men with
occur, but it is less rigid and more stimulus bound primary hypogonadotropic hypogonadism respond in
than during testosterone replacement. Thus, whereas a similar way to those with secondary hypergonado-
some degree of erectile response to erotic stimuli can tropic hypogonadism. The reported results have been
occur without testosterone, it is necessary for a full inconsistent; Gooren (1988) compared two groups of
erection and also for a state of arousal that results in six teenage males with delayed puberty aged between
the erection continuing for a time after the sexual 14 and 18 years; one group hypogonadotropic (i.e. the
stimulus is switched off (Carani et al. 1995). This primary problem being lack of gonadotropic stimula-
can be seen as a further manifestation of the central tion of the testes), the other hypergonadotropic (i.e.
excitatory tone. the primary problem is failure of the testes to respond
These studies of testosterone withdrawal and to gonadotropic stimulation). The hypogonadotropic
replacement reviewed so far, involved hypogonadal group, while responding to testosterone replacement
men in Europe or North America. In the evaluation in a similar way in their erectile and ejaculatory
of a new androgen, 7a-methyl-19-nortestosterone or responsiveness, showed less increase in sexual
MENT, a similar placebo-controlled design was used, interest and frequency of sexual activity, and less
involving two small groups of hypogonadal men: one positive change in mood and energy than the

91
Chapter 5: The behavioral correlates of testosterone

hypergonadotropic group. A crucial issue is whether a However, a number of studies have been published
hypogonadal male has gone through normal pubertal that give us information about the behavioral effects of
development before the onset of hypogonadism. If he increasing testosterone in eugonadal men. Anderson
has not then there may be some residual abnormality et al. (1992) carried out a placebo-controlled evaluation
of pubertal development that results in a deficient or of 31 healthy men who were involved in an ongoing
altered brain response to testosterone. More research efficacy trial of hormonal male contraception. They
is needed on this issue. were divided into two groups. One group had testoster-
one enanthate im injections of 200 mg weekly for eight
weeks; in the other group weekly injections of placebo
5.3.1.2 Experimental reduction of testosterone were given for four weeks followed by four, weekly
in eugonadal men injections of testosterone enanthate. Neither group
In a study of normal men, aged 2040, Bagatell et al. showed any change in measures of sexual interactions
(1994a) lowered endogenous testosterone by means of with partners or masturbation. Both groups, however,
a GnRH antagonist, NalGlu, on its own or in com- showed an increase in subscale 2 of the Sexual Experi-
bination with 50 or 100 mg of intramuscular (im) ence Scale (Frenken and Vennix 1981) during testos-
testosterone enanthate or a placebo, each in a group terone administration, but this did not happen with
of 9 or 10 men. This manipulation lasted for six weeks placebo. This subscale is an index of sexual interest
for each group. There was a clear reduction of sexual independent of interaction with a partner. Bagatell
interest and activity when testosterone was reduced et al. (1994b) administered testosterone enanthate
with NalGlu alone, consistent with the changes 200 mg im to 19 eugonadal men. There were no effects
reported in withdrawal of testosterone replacement on frequency of sexual activity, including masturba-
from hypogonadal men reviewed above. However, tion, though there was a non-significant increase in
these changes were avoided with even the lowest dose reports of spontaneous erections. Yates et al. (1999)
of testosterone replacement (50 mg testosterone compared three dose levels of testosterone in eugona-
enanthate weekly), which resulted in testosterone dal men over a 14-week period, using a placebo con-
levels well below the baseline levels. It is possible that trol. They found no increase in frequency of orgasm or
if these low testosterone levels had been sustained daily ratings of sexual interest. Buena et al. (1993)
over a longer period there may have been negative recruited two groups of healthy men, aged 1849 years,
consequences. Comparable findings were reported by and suppressed testicular function with a GnRH analog
Behre et al. (1994), who used a different GnRH antag- (leuprorelin). One group was then given doses of tes-
onist, cetrorelix. Overall, these findings indicate that tosterone replacement to achieve levels in the lower
for most men the normal range of testosterone is part of the normal range, and, in the other group, to
above that required for testosterone-dependent sexu- achieve levels in the high part of the normal range.
ality. We will return to possible explanations for this They were assessed for nine weeks on this regime
seemingly paradoxical fact later in this chapter. and, towards the end of this period, their NPT
The effects of increasing testosterone levels was assessed in a sleep laboratory. The two groups
above the normal range for extended periods has did not differ in measures of sexual interest or activity
become an issue with the recent interest in hormo- or in NPT (although rigidity of the NPT was not
nal contraception for males. Although relatively assessed).
high doses of exogenous testosterone suppress There is therefore some inconsistency in this
spermatogenesis, mainly by suppressing FSH, these limited literature that may reflect differing levels of
effects are not well sustained over time, and side- testosterone attained, and different methods of assess-
effects are significant. Attention is therefore being ing their effects. It remains possible, however, that
given to combinations of testosterone, usually in a there is a normal range of testosterone which may
slow-release form, and a progestogen (e.g. dieno- vary to some extent across men, and that varying
gest). Progestogens, while effective in suppressing testosterone levels within the individuals normal
spermatogenesis, also have side-effects, and as range will produce no sexual effect, with levels below
yet no fully satisfactory regime for hormonal male the individuals range resulting in impairment, and
contraception has been established or approved with levels above his range, an increase most predict-
(Manetti and Honig 2010). ably in sexual interest. Thus we can reasonably

92
Chapter 5: The behavioral correlates of testosterone

conclude that levels of testosterone in this normal secretion. In one group this was countered by testos-
range are necessary for the sexuality of men, which, terone replacement from the start (n = 17); the other
given its fundamental role in reproduction, is not group (n = 17) received leuprorelin alone for four
likely to show much genetic variability. weeks and then placebo was added. Mood was
assessed by the BDI (Beck et al. 1961) plus daily
5.3.2 Testosterone, mood and self-ratings. Three men in the leuprorelin plus
placebo group showed an increase in BDI score to 7
aggression in men or higher, and one of these men met DSM-IV (Diag-
5.3.2.1 Depression nostic and Statistical Manual of Mental Disorders)
Although reduction of sexual interest is more likely criteria for depressive disorder, his BDI score rising
when testosterone levels are below the normal range, from 0 at baseline to 14. The authors concluded that
there is some evidence that depressed mood also may short-term hypogonadism is sufficient to precipitate
occur. This has received the most attention in older depressive symptoms in only a small minority of
men. Barrett-Connor et al. (1999) studied 856 men, younger men. The predictors of this susceptibility
aged 50 to 89, who were attending a medical clinic. remain to be determined. Whether this susceptibility
Depressed mood was assessed using the Beck Depres- increases with age remains uncertain.
sion Inventory (BDI; Beck et al. 1961). Scores for the
BDI were significantly and negatively associated with 5.3.2.2 Aggression
bioavailable testosterone and also, though less signifi- The idea that testosterone is the aggression hor-
cantly, with DHT. Twenty-five of the men were diag- mone, accounting for why men are more aggressive
nosed as clinically depressed, and their bioavailable than women, is a widely held myth often exploited by
testosterone was 17% lower than the remainder of the the media. Most of this chapter has focused on sexual
sample (p < 0.01). Shores et al. (2005) assessed 748 behavior, and we have seen an unmistakable link
men aged 50 years or older without any previous between testosterone and male sexuality, although
history of diagnosed depressive illness. Men who at the precise nature of this link remains uncertain. With
the outset had low testosterone levels were signifi- aggression, we are faced with a great deal of inconsist-
cantly more likely to become depressed over the next ent evidence that is mainly based on attempts to cor-
two years, independently of medical morbidity. relate circulating levels of testosterone with personality
Almeida et al. (2008), in an Australian community characteristics related to aggression.
study, assessed 3987 men aged 71 to 89 years. Of these Harris (1999) provides an extensive review of the
men, 203 (5.1%) were clinically depressed and they literature that vividly demonstrates this inconsistency.
were significantly more likely to have total and free Various possibilities have been proposed. Testoster-
testosterone levels in the lowest quintile. one may influence the aggressive personality either
Amiaz and Seidman (2008) reviewed the relevant directly, as suggested by Christiansen and Knussman
literature on testosterone and depression in men. (1987), or indirectly through other aspects of the
They pointed out that in most of the earlier controlled personality relevant to aggression. Thus testosterone
studies of testosterone replacement in hypogonadal may be related to irritability (Brown and Davis 1975);
men, which focused on sexual effects, depressive to the individuals level of frustration tolerance
mood was not systematically or adequately assessed. (Olweus et al. 1980; 1988); to dominance (Gray
In several more recent randomized controlled trials of et al. 1991) or to impulsivity (Gladue 1991). Caprara
testosterone replacement, which have focused more et al. (1992) proposed that irritability plays a crucial
on depressed mood, the results have been inconsist- role as a bridge construct between emotional and
ent. They concluded that there was no pervasive influ- cognitive components of aggression.
ence of testosterone on mood, but there might be a Huesmann and Eron (1989) emphasized the
subgroup of depressed men who would benefit from developmental aspects of aggression. They proposed
testosterone therapy. This had been suggested by that severe, antisocial aggression usually emerges
Schmidt et al. (2004). In an experimental study, 34 early in life, and that certain genetic characteristics
young, healthy volunteers (aged 23 to 46) had their and perinatal or traumatic events predispose a child
endogenous testosterone levels reduced by leuprore- to develop aggressive habits. Once established, these
lin, a GnRH analog that downregulates LH and FSH cognitive structures may be extremely resistant to

93
Chapter 5: The behavioral correlates of testosterone

change. Through elaborative rehearsal of specific Hines (2004) made an important point about how
scripts, more general abstract scripts for social behav- testosterone might influence behavior by increasing
ior are formed which are equally resistant to change. arousal: In terms of physiological states, it is thought
However, testosterone was not mentioned by Huesmann that increased arousal strengthens behavioral procliv-
and Eron (1989). We might speculate that testosterone ities that already exist, and so can amplify pre-existing
has an organizing role, or may activate established inclinations to aggression. [. . .] arousal can some-
patterns of behavior, of the kind they described. Not times be mislabeled as a different emotion, including
surprisingly, in the conclusion to her review, Harris anger or hostility. [. . .] arousal, either high or low,
commented: . . .it is still not clear what relationship can be an aversive state and can increase the likeli-
testosterone has with aggression! hood of aggression in a manner similar to that pro-
A review of the literature on androgens and duced by situational factors.
aggression across species was provided by Hines In the previous section we saw central arousal as
(2004). Whereas the relationship between testoster- a complex state that we experience as sexual arousal.
one and aggression may be more clear-cut in other However, the specifically sexual components are the
non-primate species, gender difference is variable genital response, and information processing that is
across species, and the aggressive potential of female focused on sexual meanings. The central arousal that
mammals has been underestimated. Meta-analytic occurs is not necessarily different to other types of
data from humans show that gender difference in arousal, although there is some suggestion that it
physical aggression is moderate (effect size, d = 0.50), might be influenced by testosterone receptors (ARs)
larger in children (d = 0.58) than in college students in the noradrenergic system that, in general, deter-
(d = 0.27) (Hyde 1984). This underlines the import- mines central arousal in a non-specific way. What is
ance of the developmental process, with presumably not clear is whether these ARs allow an arousal
an early organizational effect of testosterone on response that is specific to sexual situations, or
prepubertal aggressive behavior. There may also be whether they allow testosterone to have a general
a second organizational phase with puberty and asso- arousal effect not restricted to sex.
ciated brain development, though as yet it is not So far, the literature has suggested there is a small
clear whether this moderates, or in any way influ- minority of men who might show aggressive
ences the gender difference seen previously in responses to changes in testosterone within the
childhood. normal range. We have no clear evidence of aggres-
Book et al. (2001) reported a meta-analysis of sion in hypogonadal men but no indication that they
published studies in humans that showed the average become more aggressive with testosterone replace-
correlation between plasma testosterone levels and ment. Let us, therefore, focus on the six controlled
aggression to be small (r = 0.14). They suggested that studies that have assessed the effects of supraphysio-
this could also be an overestimate because of under logical levels of testosterone on mood and aggression
reporting of negative findings. in eugonadal men.
In both human and non-human primates, experi- In the placebo-controlled evaluation of sexuality
ence of success in dominance encounters can cause a and mood in 31 healthy men by Anderson et al.
rise in testosterone, at least temporarily. Furthermore, (1992), reported earlier, daily ratings of various
in human studies, experience (e.g. of winning versus aspects of mood were collected; these included
losing) and internal states (motivation and anticipa- tense, energetic, unhappy, irritable, ready to
tion of certain types of competition) can influence fight, easily angered. None of these showed any
testosterone levels (e.g. Neave and Wolfson 2003). change with supraphysiological levels of testosterone
However, the functional relevance of such short-term compared to baseline or placebo.
increase in circulating testosterone is not clear. It is Tricker et al. (1996) used a double-blind,
sometimes assumed that it will increase behaviors placebo-controlled design, and administered 600 mg
relevant to aggression or dominance, but there is of im testosterone enanthate per week. The study was
little or no evidence to support this assumption divided into a 4-week baseline period, a 10-week
(Hines 2004). Overall, Hines concluded, the evidence treatment period and a 16-week recovery period.
points more to greater dominance resulting in higher Forty-three eugonadal men, 1940 years of age, were
testosterone than vice versa. randomized to one of four groups: Group 1, placebo,

94
Chapter 5: The behavioral correlates of testosterone

no exercise; Group 2, testosterone enanthate, no testosterone cypionate. In the placebo condition,


exercise; Group 3, placebo plus exercise; Group 4, equivalent amounts of the sesame oil vehicle were
testosterone enanthate plus exercise. Exercise con- injected. Assessment of mood and aggression
sisted of thrice-weekly strength-training sessions. involved the Young Mania Rating Scale (YMRS;
The Multidimensional Anger Inventory (MAI; Seigel Young et al. 1978), the 24 item Hamilton Depression
1986), which includes five different dimensions of Rating Scale (Hamilton 1960) and the Buss and Perry
anger (inward anger, outward anger, anger-arousal, Aggression Questionnaire (Buss and Perry 1992),
hostile outlook and anger-eliciting situations), and a together with the Symptom Checklist-90-R
Mood Inventory (MI), were completed by subjects (Derogatis 1994). In addition, daily diaries of manic
before, during and after the 10-week intervention. and depressive symptoms, and how much he liked
The mans significant other (e.g. spouse, or parent) his current state, were completed by each participant,
answered the same questions about the subjects and a similar weekly diary was completed by a sig-
mood and mood-related behavior. There were no nificant other. During the second year of the study
significant changes in any of the measures during an additional measure was added: the Point Subtrac-
the treatment period, and no differences in these tion Aggression Paradigm (PSAP; Kouri et al. 1995).
variables between exercising and non-exercising or Each participant was seated in a booth equipped with
between placebo and testosterone enanthatetreated a monitor screen and was told that he was playing
subjects. The authors concluded that supraphysiolo- against an unseen male opponent, which was actually
gical doses of testosterone, when administered to a computer. The participant could accumulate points
normal men in a controlled setting, do not increase on the screen exchangeable for money by pressing
angry behavior. They did not, however, exclude the one button, or he could deprive his opponent of
possibility that high doses might provoke angry points by pressing another button. During the ses-
behavior in men with a particular susceptibility to sion, the opponent provoked the participant by ran-
angry responses. domly depriving him of points. The participants
In the Yates et al. (1999) study reported earlier, aggression score represented the total number of
mood was monitored throughout the study using the points that he subtracted from his opponent in retali-
BussDurkee Hostility Inventory (BDHI; Buss and ation to this provocation. Subjects who realized that
Durkee 1957), the Brief Psychiatric Rating Scale the opponent was actually a computer were excluded
(BPRS: Overall and Gorham 1962), the Modified from the analysis of this variable.
Mania Rating Scale (MMRS; Blackburn et al. 1977) Manic scores on the YMRS increased significantly
and the Hamilton Depression Rating Scale (HDRS; with testosterone treatment (p = 0.002). However, this
Hamilton 1967). Only minimal effects on these meas- effect resulted from a few participants reporting
ures of mood and behavior were observed during the marked manic-type symptoms, whereas the majority
14-week testosterone administration and withdrawal showed little change. No participant reported violent
phases for those who completed the study, and there behavior while receiving testosterone, but several
was no evidence of a dose-dependent effect on any of described instances of uncharacteristic aggressiveness;
the above measures. There were also no effects on one participant was withdrawn from the study after
psychosexual function. One participant on 500 mg the fifth week because he became alarmingly hypo-
of testosterone developed a brief state of agitated manic and aggressive. Aggression scores on the
and irritable mania, and was therefore withdrawn PSAP increased significantly with testosterone treat-
from the study. ment (p = 0.03) in spite of the smaller number of
Pope et al. (2000) recruited 56 men, aged 20 to 50 participants in this analysis (n = 27). Again, however,
(77% in the 2029 year range), into a randomized, this effect depended on a few showing marked
placebo-controlled crossover study. Participants were changes whereas most participants showed little
seen weekly for 25 weeks. This covered six weeks for change. The manic and liking scores on partici-
treatment 1, six weeks for washout, six weeks for pants daily diaries also showed a significant testoster-
treatment 2 and seven weeks for final washout. In one treatment effect. Diaries from significant others
the active treatment, the first two, weekly injections did not show a significant testosterone effect;
contained 150 mg, the third and fourth weeks, 300 although, at the end of the testosterone treatment
mg, and the fifth and sixth week, 600 mg of period, the diary manic scores from participants and

95
Chapter 5: The behavioral correlates of testosterone

their significant others correlated significantly days. There were subtle and statistically significant
(Spearman r = 0.59; p < 0.001). (p < 0.05) increases in positive mood (euphoria,
OConnor et al. (2004) used a double-blind, energy and sexual arousal), negative mood
placebo-controlled crossover design in a study of 28 (irritability, mood swings, violent feelings and hostil-
healthy eugonadal men (mean age 32.3 years). This ity), although no aggressive behavior, and some cog-
involved three eight-week periods; the middle period nitive impairment (distractibility, forgetfulness and
being washout. A single im injection of testosterone confusion). As with the previous studies, these group
undecanoate 1000 mg was compared to a single pla- changes concealed considerable individual variability.
cebo injection, with randomization to one of two One subject, for example, had an acute manic episode.
sequences (i.e. testosterone undecanoate placebo, These variable reactions were not predicted by base-
or placebo testosterone undecanoate). Following tes- line characteristics or family psychiatric history.
tosterone undecanoate administration the testoster- The overall conclusion from these six controlled
one levels were substantially raised for two weeks, studies is that for the majority of eugonadal healthy
though the LH levels remained suppressed for longer. men, temporary increases of plasma testosterone to
The Profile of Mood States (POMS; McNair et al. supraphysiological levels do not increase aggressive
1992) was used to monitor mood. Various aspects of feelings or behavior. But there is a minority who are
aggression, including both feelings and behavior, affected, though in most such cases the effects have
were monitored with the Aggression Questionnaire been more manic than aggressive. Such effects are
(AQ; Buss and Perry 1992), which was also completed suggestive of a relatively non-specific arousal mech-
by the partner (Partner Aggression Questionnaire or anism, the manifestations of which may depend on
AQ-P; OConnor et al. 2001), the Aggressive Provo- personality characteristics or the particular circum-
cation Questionnaire (APQ; OConnor et al. 2001) stances at the time. It is possible that the relevant
and the Irritability subscale from the BussDurkee personality characteristics are the same as those that
Hostility Inventory (Buss and Durkee 1957). In add- result in depressive reactions to lowered testosterone
ition, the Rathus Assertiveness Schedule (Rathus levels, as discussed above. It remains uncertain as to
1973) and the State Self-Esteem Scale (Heatherton what extent these short-term effects of supraphysio-
and Polivy 1991) were used. There were significant logical plasma testosterone levels are based on the
increases in anger/hostility scores for the first two same hormone-behavior mechanisms that result from
weeks after testosterone undecanoate injection, but normal levels.
at levels that stayed within the normal range for US
college samples. This was accompanied by a signifi- 5.3.2.3 Testosterone and sleep
cant reduction in fatigue/inertia. There was no If arousal is the essence of testosterone effects in the
increase in aggressive behavior. brain, how might it affect sleep? It has been recog-
The final controlled study is unique in that testos- nized for some time that testosterone administration
terone was increased for only three days, and hence can induce sleep apnea (e.g. Sandblom et al. 1983).
allows us to assess short-term effects of testosterone Conversely, sleep apnea is known to reduce testoster-
on mood. Su et al. (1993) reported a fixed-order, one as a result of dysfunction of the pituitary-gonadal
placebo-controlled, double-blind study. Twenty axis (Luboshitzky et al. 2002). Sleep deprivation also
healthy men (mean age 27.5 years), with no prior reduces circulating testosterone levels in healthy men
history of anabolic steroid use, were admitted to the (e.g. Corts-Gallegos et al. 1983).
National Institute of Mental Health inpatient research Leibenluft et al. (1997) explored the effects of
ward for 14 days. This therefore involved another testosterone on sleep by inducing transient hypogo-
unique feature; all subjects were kept in the same nadism with leuprorelin, a GnRH analog. This was
environment during their participation in the study. given for three months to 10 normal healthy male
After two days settling in, they experienced four volunteers with an average age of 29 years. In the
consecutive three-day conditions. The first three days second and third months, at two week intervals, either
were a placebo baseline, then oral methyltestosterone testosterone enanthate 200 mg or placebo were given
40 mg daily (the normal prescribed dose) was given in a double-blind, randomized order, crossover
for three days, followed by methyltestosterone 240 mg design. Although no differences in timing or duration
daily for three days, followed by placebo for three of sleep were observed, there was a significant

96
Chapter 5: The behavioral correlates of testosterone

reduction in the proportion of stage 4 sleep, and explain this minority response, and, if at all pos-
reduction of 24-hour prolactin levels, in the hypogo- sible, to assess its heritability.
nadal state compared with testosterone replacement.
The authors concluded that testosterone has rela-
tively specific and discrete effects on sleep and hor- 5.3.3 Summary of evidence in the male
monal rhythms in men. There is consistent evidence of a relationship between
Liu et al. (2003) carried out a placebo-controlled testosterone and both sexual interest and NPT in
crossover evaluation of the effects of testosterone younger men. This suggests that the main central
administration on sleep in older men. In the active effects of testosterone are on central arousal associ-
phase, three, weekly im injections of Sustanon (a ated with response to sexual stimuli (Bancroft 1995).
mixture of three testosterone esters) were given: As a consequence, thoughts about sex, accompanied
500 mg in the first week, and 250 mg in the second by central arousal, are experienced as manifestations
and third weeks. In the placebo condition, weekly of sexual interest. The association with NPT is
injections of the excipient oil were given. In between suggestive of a testosterone-dependent excitatory
treatments there was an eight-week washout period. tone based on central arousal mechanisms, which is
Sleep and breathing were assessed by polysomnogra- allowed expression through penile erection when the
phy at baseline, and at the end of each treatment inhibitory tone maintaining the normally flaccid
period. Seventeen healthy men over the age of 60 penis is reduced or switched off during REM sleep.
participated. Total sleep time was reduced by an The relevance of testosterone to erectile function
average of one hour, and disrupted breathing is more subtle or complex. There is recent evidence of
increased; both significant differences (p < 0.05). testosterone-dependent mechanisms in the spinal
There was, however, no evidence of change in upper cord and penis that are relevant (Montorsi and Oettel
airway dimensions, making it unlikely that the 2005). We can assume that testosterone-dependent
sleep disruption was due to airway obstruction. The arousal mechanisms will augment erectile response.
question arises of whether this negative effect of But other mechanisms, including cognitive process-
testosterone administration is more likely in older ing of sexual stimuli, as well as age and disease-related
men. In the six studies of supraphysiological testos- increases in erectile inhibitory tone, which are not
terone administration in eugonadal men, all less than testosterone dependent, have an equal or greater
60 years of age, reviewed above, negative effects on influence on erectile response.
sleep were only mentioned in two reports. Su et al. Whereas the picture is comparatively clear during
(1993) included a rating of insomnia in their daily young adulthood and midlife, developmental pro-
diaries. They found a dose-related trend towards cesses involved both around puberty and with aging
more insomnia with testosterone administration are less well understood. With the recent awareness of
(p < 0.1). As each dose was only for three days, important brain development during the second
one wonders whether this may have become more decade, it is possible that testosterone is having an
marked with longer usage. Yates et al. (1999) organizing as well as an activating effect around
described one participant who withdrew from the puberty. With aging, there are complex changes in
study because of an acute onset of distress, associated testosterone receptor number and sensitivity, as well
with marked irritability and sleep-onset insomnia. as other changes both centrally and peripherally,
Clearly, testosterone is related to sleep in ways that which makes the physiologic function of testosterone
may be important in states of testosterone deficiency more obscure.
and during testosterone replacement. But as yet we The concept of a threshold below which androgen
have an incomplete and somewhat confused picture deficiency occurs, and above which increasing andro-
of this relationship. gen levels have little effect, is fairly consistently sup-
While the research reviewed here has indicated ported by the evidence. However, our ability to
that a minority of men are susceptible to the hyper- identify the threshold level remains limited. The
arousing effects of testosterone administration, it level is likely to vary across individuals, and it may
has not yet enabled us to identify relevant charac- possibly change with age and exposure to sustained
teristics of these responders. Future research should and different levels of testosterone. At present, we can
focus on potential predictors that might help to only speculate on this issue. A number of studies have

97
Chapter 5: The behavioral correlates of testosterone

suggested that subtle effects may arise from increasing pleasure and/or orgasm, is it desire for emotional
testosterone to supra-threshold levels. intimacy or closeness to ones partner, is it desire to
We have little systematic evidence on the timing give ones partner pleasure, or is it a combination of
of testosterone effects on sexual responsiveness. The these? And how do these vary across women, and
hormone replacement studies discussed above suggest within the same woman dependent on context, and
that, after a period of testosterone withdrawal, it is at different stages of life?
several days or even two-to-three weeks before In the male, genital response, or penile erection, is
replacement testosterone has its maximum effects. obvious to him and hence there tends to be a high
On the other hand, short-term administration of tes- correlation between sexual desire and awareness of
tosterone to eugonadal men has apparently produced some degree of erection in men. The homologous
effects, albeit subtle, within three days (e.g. Carani form of genital response in the woman is clitoral
et al. 1990; Su et al. 1993). Further research is tumescence, and this is less obvious to her and hence
required on this basic issue. less central to her experience of sexual arousal.
Whereas testosterone effects on sexuality are rela- Women, in their experience of sexual desire, conse-
tively universal in men, testosterone effects on mood quently focus more on whether they are feeling
and aggression, observed only in a minority of men aroused in a more subjective sense (Graham 2010).
studied, suggest that some men are more sensitive to In women there are two aspects of genital response
such effects. which are uniquely female; the increased blood flow
and pulse amplitude in the vaginal wall (as measured
5.4 Activational effects of by vaginal pulse amplitude (VPA; Laan and Everaerd
1995), and the reduced sensitivity to pain from
testosterone and behavior in vaginal or cervical stimulation (Komisaruk and San-
adult women sone 2003), both of which are crucial to allow vaginal
The role of testosterone, at least in relation to sexual- penetration without discomfort. But these two mech-
ity, is better understood in men than in women, anisms may not otherwise contribute to the womans
despite the fact that there is much more evidence in experience of sexual arousal or desire.
the literature relating to hormones and female sexual- Most women experience orgasm, but the hormo-
ity, and more opportunities to study variations of nal regulation of this complex and ill-understood
hormones in the female (e.g. menstrual cycle, lacta- phenomenon remains unclear in both women and
tion, menopause). This reflects the complexity and men. Seminal emission in men, which is linked to
greater inconsistency of the evidence in women. Later orgasm in some way that is also not well understood,
in this chapter, direct comparisons of the role of is testosterone dependent. There is no equivalent of
testosterone in men and women will be made. seminal emission in women.
Paradoxically, in research on the sexuality of The relevance of testosterone and other androgens
women, more attention has been paid to testosterone to womens sexuality will be reviewed under the
than to estradiol. As mentioned earlier, one crucial following headings: (1) circulating androgen levels
and unresolved issue is the extent to which testoster- in women; (2) iatrogenic lowering of androgens and
one produces its effects, in both male and female, by (3) exogenous androgen administration.
conversion to estradiol.
5.4.1.1 Circulating androgen levels and their relation
5.4.1 Sexual desire and response to the sexuality of women
As in men, the essence of womens sexual desire is Plasma total testosterone in women is on average less
incentive motivation linked to processing of sexually than 3.5 nmol/l; i.e. around 10% of levels in the male.
relevant information, with varying degrees of central Given the problems with assaying low levels of testos-
arousal and genital response. However, there is little terone, there is no clear bottom of the normal range.
evidence of what is being desired in women, and there Consistent with that is the absence of any recogniz-
may be important gender differences, or at least able testosterone deficiency in women, except when
greater variability in women than in men (Bancroft there has been an iatrogenic lowering (see below).
and Graham 2011). In particular, is it desire for sexual Dehydroepiandrosterone and DHEAS are present in

98
Chapter 5: The behavioral correlates of testosterone

the plasma at levels as high as in men, at least in explanation for this is that the establishment of a
premenopausal women. sexual problem, with its various repercussions in
To what extent do plasma levels of testosterone the relationship, may serve to obscure subtle hor-
relate to the sexuality of women? Davis et al. (2005) monebehavior relationships. It would therefore be
asked a community-based sample of 1021 Australian interesting to know whether there was any correlation
women, aged 18 to 75 years, to complete the Profile of between testosterone levels and PFSF scores across
Female Sexual Function (PFSF; Derogatis et al. 2004), the large non-problem component of the Davis et al.
and a measure of general well-being. One fasting sample.
morning blood sample was collected. In premenopau- Few studies have investigated testosterone levels in
sal women this was collected between day 8 of the women presenting specifically with problems of low
cycle and onset of the next menses. (This is likely to sexual desire. Stuart et al. (1987) and Schreiner-Engel
obscure the modest but significant mid-cycle rise in et al. (1995) found no difference in testosterone levels
testosterone that typically occurs.) Blood was assayed between women with low sexual interest and controls;
for testosterone (total and free), androstenedione and on the other hand, Riley and Riley (2000) found a
DHEAS. lower Free Androgen Index (FAI) in women com-
The PFSF provides seven domain scores (desire, plaining of lifelong absence of sexual drive compared
arousal, orgasm, pleasure, sexual concerns, respon- to controls. These were all small studies, with
siveness and self-image). These scores were not nor- numbers in each group ranging from 11 to 17.
mally distributed in this sample, and were related to Compared with men, there are natural variations
age. The sample was therefore divided into two age of testosterone in women of reproductive age that
groups (younger, 1844 years, n = 339; older, 4575 warrant attention.
years, n = 646). For each domain these two age groups
were divided into those with low scores (zero for the The menstrual cycle
older; the lowest 5% for the younger group) and the Testosterone levels typically rise during the follicular
rest. The low scorers and the rest for each domain phase and are at a maximum approximately for the
were then compared for plasma levels of androgens. middle third of the cycle, declining during the final
Neither total nor free testosterone discriminated third of the cycle to reach the lowest levels for the first
between these groups for any domain of sexual func- few days of the next follicular phase. In some women,
tion, in either the younger or older women. Dehy- this mid-cycle rise in testosterone is a more discrete
droepiandrosterone sulfate, however, was periovulatory peak. In a study of 37 women with
significantly lower in the low scorers in three normal ovulatory cycles, testosterone levels were
domains for each age group. For the younger group, highest during the mid-third of the cycle (Bancroft
the three domains were desire, arousal and et al. 1983). In the 21 women who masturbated,
responsiveness; for the older group: arousal, frequency of masturbation was significantly correl-
responsiveness and pleasure. In addition, ated with mid-cycle testosterone levels; there was no
androstenedione was significantly lower for the older correlation between testosterone and frequency of
low responders in the pleasure domain. sexual activity with partner. There is now an extensive
This study by Davis et al. (2005) is by far the literature on variations of sexual interest and behavior
largest of its kind. However, their comparison of the through the menstrual cycle, though the findings are
low scorers on the PFSF with the rest leaves us with somewhat inconsistent (see Hedricks 1994 for a
some unanswered questions. Many of the women in review). Sexual activity tends to be lowest during the
the low scorer groups may have seen themselves as menstrual phase, and sexual interest to be highest
having a sexual problem. In an early and small during the follicular phase or around ovulation,
study of oral contraceptive users, we found that though there is considerable individual variability.
women who defined themselves as having a sexual Although mid-cycle increases in testosterone could
problem did not show any correlation between tes- be contributing to this pattern, other hormonal and
tosterone levels and a measure of sexual interest; non-hormonal explanations also have to be
whereas women who did not regard themselves as considered.
having a sexual problem showed a significant correl- Lactation is associated with a lowering of plasma
ation (Bancroft et al. 1980; see below). One possible testosterone as well as estradiol, and often with

99
Chapter 5: The behavioral correlates of testosterone

reduced sexual interest. One small study (Alder et al. and the importance of fertility, amongst other things,
1986) showed that breast-feeding women with low influence the picture. As yet, no predictable associ-
sexual interest had significantly lower plasma testos- ation between the age-related decline in androgens
terone levels than breast feeders with normal sexual and changes in the sexuality of older women has been
interest; however, this finding still awaits replication. demonstrated.

5.4.1.2 Aging and the menopause 5.4.1.3 Effects of iatrogenic lowering of testosterone
Adrenal androgens, particularly DHEA and DHEAS, in women
show an age-related decline in women over a rela-
There are three principal iatrogenic procedures that
tively wide age span (Orentreich et al. 1984; Bancroft
happen to reduce testosterone in women: oral contra-
and Cawood 1996; Sulcov et al. 1997; Burger et al.
ceptive use, use of antiandrogens, and bilateral
2000; Davison et al. 2005). This was most convin-
ovariectomy.
cingly demonstrated in a study of ovariectomized
women where ovarian androgens could not obscure
the picture (Crilly et al. 1979). Ovarian androgens Oral contraceptives
present a more complex picture. Decline in testoster- It has been known for some time that oral contracep-
one starts a few years before the menopausal transi- tives reduce free testosterone (Jung-Hoffman and
tion, probably due to a reduction in the mid-cycle rise Kuhl, 1987; Van der Vange et al. 1990; Janaud et al.
of testosterone (Roger et al. 1980; Zumoff et al. 1995; 1992; Darney 1995; Thorneycroft et al. 1999; Boyd
Mushayandebvu et al. 1996). The menopause per se et al. 2001). Mid-cycle rises in testosterone and
has less predictable effects on ovarian androgens. In a androstenedione are blocked by suppression of ovu-
longitudinal study of 172 women who made the tran- lation and the normal pattern of gonadal steroid
sition from premenopausal to postmenopausal during production. In addition, the estrogen in the oral con-
the seven years of the study (Burger et al. 2000), total traceptive increases SHBG levels and hence reduces
testosterone did not change, whereas free testosterone the free testosterone available. There is more limited
increased from pre- to postmenopausal. In the large evidence of a reduction in DHEA and DHEAS
Australian cross-sectional study, described above, a (Coenen et al. 1996; Graham et al. 2007).
steady decline in testosterone (both total and free), In spite of this consistent evidence, it is striking
androstenedione and DHEAS was found across age how little attention has been paid to the possible
groups, with menopausal status having no influence effects of this decline in androgens on womens sexu-
on this decline (Davison et al. 2005), and there was no ality (Bancroft and Sartorius, 1990; Davis and Cas-
age-related change in SHBG. tano 2004). In an early study (Bancroft et al. 1980), we
The function of the interstitial cells of the ovary compared 20 established oral contraceptive users
changes with the menopause. In premenopausal complaining of sexual problems that they attributed
women, gonadotropic stimulation of the interstitial to the oral contraceptive, with 20 oral contraceptive
cells is regulated by the negative feedback of ovarian users without sexual problems, matched for oral con-
steroids. However, the rise in LH that accompanies traceptive and age. The total testosterone levels were
the menopausal transition, resulting from the reduc- low in both groups (free testosterone was not esti-
tion in estrogen-induced negative feedback, may mated) and did not differ significantly. As mentioned
stimulate the interstitial cells to produce testosterone earlier, we found a significant correlation between our
and androstenedione, sometimes excessively. In add- measure of sexual interest (frequency of sexual
ition, factors such as body weight and insulin resist- thoughts) and the testosterone level in the no prob-
ance influence this ovarian androgen production (see lem group (r = 0.65; p < 0.01) but not in the prob-
Cawood and Bancroft (1996) for a brief review of the lem group (r = 0.02; non-significant). A significant
evidence). Some researchers, however, have ques- correlation between plasma testosterone and sexual
tioned whether the postmenopausal ovary is a signifi- interest was found in two other studies of oral con-
cant source of androgens (e.g. Couzinet et al. 2001). traceptive users (Bancroft et al. 1991; Alexander and
The variability of womens sexuality, striking Sherwin 1993). These three studies of women on oral
enough in younger women, becomes even more contraceptives, and without sexual problems, are so
marked in older women, when psychosocial factors far the only studies to show significant correlations

100
Chapter 5: The behavioral correlates of testosterone

between plasma testosterone and sexual interest in a decline in testosterone after starting the oral contra-
women. Thus these correlations are with testosterone ceptive was associated with low levels of sexual interest.
levels in the lower part of the physiological range. But there were other women who experienced a sub-
This led Alexander and Sherwin (1993) to postulate stantial decline in free testosterone and no apparent
a threshold effect in women, comparable to that in reduction, or even an increase in sexual interest. We
men, but at a much lower level of plasma testosterone; have to keep in mind the varied psychological effects
i.e. correlations between plasma testosterone level and that an effective contraceptive may have on a womans
measures of sexual interest will only become apparent sexuality. In some women the freedom from concern
when the testosterone levels are around or below this about pregnancy may have a sexually liberating effect.
threshold level, which is presumably in the lower part We thus have to balance the effects of such inhibition
of the normal range. reduction with the possible negative effects of testos-
Most of the relevant literature on oral contracep- terone reduction on sexual interest. Cultural factors
tive use and sexuality has involved cross-sectional influencing womens sexuality also need to be
studies, where women who experienced negative considered.
sexual effects are likely to have discontinued oral con- A range of factors may contribute to the complex
traceptive use, and hence selected themselves out of effects of oral contraceptives on sexuality, but the
the study. Recently, we reported the first study to possibility that a substantial minority of women are
measure change in free testosterone and in measures dependent sexually on testosterone, and experience
of sexuality and mood in 61 women starting on oral negative sexual changes, particularly in sexual inter-
contraceptives (Graham et al. 2007). Sexuality was est, because of the oral contraceptiveinduced reduc-
assessed by the Interviewer Ratings of Sexual Function tion in free testosterone, remains a distinct possibility.
(IRSF), and mood by the Beck Depression Inventory In comparison to the limited evidence in relation to
(BDI; Beck et al. 1961). Highly significant reductions oral contraceptives, there is virtually no systematically
in free testosterone and DHEAS were found after collected evidence of the effects of long-acting hor-
three months on oral contraceptive, though there was monal implants (e.g. Norplant) or injections (e.g.
considerable individual variability in the degree of depot medroxyprogesterone acetate, DMPA) on
reduction. As in previous studies, there was also con- womens sexuality. Given that DMPA is used in the
siderable variability in the measures of sexuality and United States to control male sex offenders (Walker
mood. Thus the frequency of sexual thoughts, the 1978), it is disturbing that an official UK Government
principal measure of sexual interest, increased in 33% report (National Institute for Health and Clinical
of women and decreased in 23%, with the remainder Excellence; NICE 2005) recommended the use of
showing no change. On the BDI, while showing on DMPA for contraception by young women when we
average very little change, 39% of women showed have no idea what effects such long-acting methods
improvement in mood and 29% worsened. Some sup- have on womens sexuality or mood.
port was found for the hypothesis that reduction in
sexual interest (and other sexuality measures) is related Antiandrogens in women
to the reduction in free testosterone. Direct correl- Antiandrogens work by blocking androgen effects at
ations between the change scores for both behavioral the receptor and also by reducing testosterone levels
and hormonal measures were not significant, but when by means of negative feedback effects. Cyproterone
we correlated change in hormone level with the actual acetate (CPA) is an antiandrogen that has been used
level of sexuality after three months on oral contracep- for many years, at least in Europe, for treatment of
tive, we found significant correlations between reduc- androgen-dependent conditions such as acne and hir-
tion in free testosterone and two behavioral variables; sutism in women. Little attention has been given to
frequency of sexual thoughts (r = 0.35; p = 0.006), and the possible sexual side-effects of such treatment. One
percentage of occasions of sexual activity with partner study systematically assessed such effects. Appelt and
when the woman felt sexually aroused or excited (r = Strauss (1986) studied 36 women who had not had
0.28; p = 0.03). We found no significant correlations sexual problems before starting on CPA, and, of
with other aspects of sexual enjoyment or response, or these, 16 (44%) reported negative effects on their sex
involving change in DHEAS, or between hormonal life; this rose to 61% if women not in sexual relation-
change and mood scores. In some women, therefore, ships were excluded.

101
Chapter 5: The behavioral correlates of testosterone

Bilateral ovariectomy only group was significantly younger at the time of


With surgical removal of the ovaries, there is an surgery (40 vs. 45 years; p < 0.001). Assessment was
immediate and substantial drop in circulating andro- carried out at six months and two years post-opera-
gens. There have been a number of studies evaluating tively. There were no differences in sexual functioning
the effects of hormone replacement in women who between the two groups.
present with sexual problems after ovariectomy, and Removal of both ovaries, in producing a substan-
these are considered below. What proportion of tial reduction of ovarian androgens in most women,
women experience sexual problems as a consequence should provide us with a test of the importance of
of having their ovaries removed has received less androgens in women. The evidence reviewed above,
attention. The following studies present a somewhat however, gives us no clear or consistent answer. One
inconsistent picture. reasonable conclusion is that there is no predictable
Nathorst-Bs et al. (1993) compared the sexuality decline in sexual functioning as a result of ovariec-
of women who had had bilateral ovariectomy and hys- tomy, and, as with oral contraceptive use, a substan-
terectomy without any hormone replacement, with two tial proportion of women can experience a reduction
groups of age-matched hysterectomized women, one in androgens without obvious adverse effects on sexu-
having received ovariectomy plus estrogen replacement, ality or well-being. There are, however, a number of
the other hysterectomy alone. They found little differ- limitations to this source of evidence:
ence between the ovariectomized women with and 1. Women undergoing such surgery, whether
without estrogen replacement, about a half of whom involving bilateral ovariectomy or only
reported a decrease in sexual interest. In contrast, 82% hysterectomy, are having the surgery because of
of those with intact ovaries reported an increase in some gynecological health problem likely to have
sexual interest post-operatively. Comparable findings an adverse effect on both their sexuality and well-
were presented by Bellerose and Binik (1993). being. Taking the immediate pre-operative period
Farquhar et al. (2002) compared 57 women who as the baseline, the expected outcome of the
had hysterectomy with both ovaries removed surgery is an improvement. In contrast with a
(O&H) and 266 women who had hysterectomy woman starting on a steroidal contraceptive,
alone (H only). They found no deterioration sexu- therefore, there is the problem of a lack of a
ally in either group at six months post-operatively; suitable baseline to evaluate the effects of
however, the hysterectomy alone group reported a androgen reduction following ovariectomy.
significant increase in sexual interest and frequency of 2. Women are not randomly assigned to H only or
sexual intercourse, presumably reflecting the health O&H, and the decision is made partly on clinical
benefits of the surgery. The women who had both indications, age and/or the womans personal
ovaries removed showed no such benefits. preference. This introduces the potential for
Aziz et al. (2005) compared 217 women who had confounding individual differences.
H only and 106 women who had O&H. Estrogen 3. Studies vary in the extent to which hormone
replacement therapy was recommended for all the replacement therapy (HRT) was involved
ovariectomized women and those in the H only following surgery, and in only one of the cited
group who had menopausal symptoms. Sexuality and studies (Bellerose and Binik 1993) was there
well-being were assessed pre-operatively and at one comparison of HRT involving estrogen only and a
year post-operatively. The O&H group showed no combination of estrogen and testosterone.
change in sexuality or well-being; whereas the H only 4. None of the studies reviewed above has taken into
group showed some worsening in 3 of the 14 sexuality consideration the possible negative effects of
variables. None of the group comparisons was signifi- hysterectomy on womens sexual response. These
cant. Both groups showed improvement in well-being. have been observed to occur to a variable extent in
There were no correlations between the observed other studies of hysterectomy (Komisaruk et al.
changes pre- to one year post-operation in androgen 2011).
levels and any aspect of sexuality or well-being. 5. With the exception of the first two studies cited,
Teplin et al. (2007) compared 49 women who had results analyzed were presented as group averages.
O&H with 112 women who had H only. The H As is the case when assessing the effects of oral

102
Chapter 5: The behavioral correlates of testosterone

contraceptives, a group average indicating no improvement than the diazepam group in satisfaction
change may conceal the fact that some women with sexual intercourse, frequency of orgasm, sexual
have experienced improvement and some arousal and frequency of sexual thoughts. These
worsening. If, as the evidence from oral results raised the question of whether this was a
contraceptive use suggests, some women are positive result of testosterone or a negative effect of
dependent on testosterone for their sexuality and diazepam on the effectiveness of the counseling.
others are not, then using group averages, in Mathews et al. (1983) attempted to replicate these
which these two types of women are combined, findings with a similar design, but using placebo
will serve to obscure this distinction. instead of diazepam. They found no difference
between testosterone and placebo. Dow and Gallagher
The studies of Nathorst-Bs et al. (1993) and (1989) questioned whether the combination of
Bellerose and Binik (1993) suggest that approximately counseling with testosterone therapy might have
a half of women experience a decline in sexuality obscured the effects of testosterone therapy alone.
following ovariectomy, and the other half do not, They therefore carried out a further study, adding a
which may make it more likely that they will report third group, who received testosterone therapy
improvements from the surgery. alone. They found significant improvement in both
of the combined groups (i.e. sex therapy plus testos-
5.4.1.4 The effects of exogenous androgen
terone or placebo) at the end of treatment and at four
administration on the sexuality of women months follow-up, with no group difference, and
Testosterone administration to healthy premenopausal women significantly less improvement with testosterone
In a psychophysiologic study, Tuiten et al. (2000) alone. In each of these three studies, 10 mg or 20
explored the timing of effects of an increase in testos- mg of testosterone was administered sublingually
terone in eight healthy women without sexual prob- (Testoral; Organon) once daily. This was shown to
lems, who were given a single sublingual dose of produce substantial increases in testosterone above
testosterone in a placebo-controlled experiment. the physiologic range, for four to five hours after
There was a rapid rise in plasma total testosterone administration (Bancroft 1989).
after testosterone administration, reaching at least a Bancroft et al. (1980) explored androgen adminis-
10-fold increase in baseline levels within the first 15 tration in 15 women with sexual problems attributed
minutes, returning to baseline within 90 minutes. to oral contraceptive use. Androstenedione was the
Subjective and VPA responses to erotic films were androgen administered. This was well absorbed when
measured just before testosterone administration, 15 taken orally, and produced increases in plasma testos-
minutes after and on four further occasions 90 min- terone as well as androstenedione. A double-blind,
utes apart. An enhanced VPA response to erotic stim- placebo-controlled crossover design was used, with
uli was observed three to four hours after the peak each woman taking androstenedione and placebo,
increase in plasma testosterone, significantly correl- each for two months. Daily diary and interview
ated with an increase in subjective reports of sexual ratings were used to assess behavioral change. Signifi-
lust and genital sensations. This is the only study to cant increases in plasma testosterone occurred in the
date that assessed the effects of an acute dosage of androstenedione-treated group, but no significant dif-
testosterone on sexual responsiveness of women. This ferences in sexuality variables resulted. One woman,
indicates a latency in testosterone effect of three to however, showed a clear improvement in sexual inter-
four hours. There are no comparable data for men. est and response following the androstenedione
administration, which was subsequently replicated,
Premenopausal women with sexual problems with administration of androstenedione followed by
There was a phase in the 1970s and 1980s when the withdrawal, on three occasions following the study. It
use of testosterone as a treatment for womens sexual is noteworthy that, as a result of treatment, she
problems was explored. Carney et al. (1978) assessed differed hormonally from the other 14 women, not
the combination of sexual counseling with either tes- in terms of increased levels of testosterone, but of
tosterone or diazepam administration, for couples in estradiol and of the estradiol : testosterone ratio.
which the woman was sexually unresponsive. The Perhaps because of the largely negative findings
testosterone group showed significantly greater from these first four studies, there followed a long

103
Chapter 5: The behavioral correlates of testosterone

period with no further research of this kind. The next later study, they assessed the effects of testosterone
comparable study in premenopausal women was not administration in such women, using a placebo-con-
published until 2003 (Goldstat et al. 2003). In a trolled, randomized design, with 24 women receiving
double-blind crossover study in Australia, 31 women 300 mg testosterone daily in a transdermal patch, and
with low libido (mean age, 39.7  4.2 years) used 27 women receiving placebo, each group for 12
testosterone cream (10 mg/day) and placebo, each months (Miller et al. 2006). In comparison with the
for 12 weeks (with a 4-week washout period between). placebo group, women in the testosterone group
Testosterone administration, which resulted in showed significant improvement in mood, measured
plasma testosterone levels at the high end of the by the Beck Depression Inventory (BDI), and in sexu-
normal range, was associated with significant ality, as measured by the combined score from the
improvements in general well-being and measures of Derogatis Interview for Sexual Function (Derogatis
sexuality (Sabbatsberg Sexual Self-Rating Scale; Gar- 1997), and the subscales for arousal and behav-
ratt et al. 1995). This was followed by a larger Austra- ior/experience.
lian study (Davis et al. 2008) in which 261 Tuiten et al. (1996) studied eight young amenor-
premenopausal women, aged 35 to 46 years, who rheic women with low weight, although none had the
reported a decline in satisfying sexual events, and also diagnosis of anorexia nervosa. They showed lower
had low serum free testosterone, were randomly levels of sexual interest and activity, and lower testos-
assigned to one of three dose levels of transdermal terone levels than an age-matched comparison group
testosterone or placebo. Free testosterone levels of normally menstruating women. The amenorrheic
(normal range 3.8 to 21.8 pmol/l) were increased in group was given testosterone undecanoate, 40 mg
the three testosterone groups, although least with the daily for eight weeks, and placebo for eight weeks,
middle dose (6.4 to 10.5 pmol/l) and most with the in a double-blind crossover study. The two
high dose (7.8 to 18.7 pmol/l). All four groups showed treatments did not differ in effects of daily ratings of
an increase in satisfying sexual events, but it was only sexuality or mood. The women were also evaluated
in the middle dose group, which showed the lowest psycho-physiologically with measurement of VPA in
increase in free testosterone, that this behavioral response to erotic fantasies and erotic films. They
increase was significantly greater than in the placebo showed a significantly greater VPA response to film
group. during testosterone administration, but this effect was
Two explanations were offered to explain the lack not reflected in subjective ratings of arousal or mood.
of dose-response relationship: (1) there may be no
benefit beyond the medium dose though this Naturally postmenopausal women
doesnt explain why the high dose was less significant Although the natural menopause has little direct
in its effects than the medium dose; (2) the higher effect on androgen levels (see Section 5.4.1.2), estro-
dose may have produced more side-effects which gen replacement (HRT) for menopausal symptoms
could counteract positive effects. There is a third increases SHBG and consequently reduces free testos-
explanation that was not considered. If it is the case terone. Conversely, testosterone administration will
that only a proportion of women are sensitive to the reduce SHBG levels and also, because of competitive
behavioral effects of testosterone, then there may have binding with SHBG, increase the amount of free
been more of these women in the middle-dose group. estradiol. Hence, it is of some interest to evaluate
It is noteworthy that there were more oral contracep- the effects of adding testosterone to the estrogen
tive users in the middle-dose group. As with most of replacement.
the earlier literature, this study does not attempt to Burger et al. (1987) reported on 20 postmenopau-
identify characteristics of testosterone-sensitive sal women whose menopausal symptoms had been
women, and results were presented only as group relieved by oral estrogen replacement but whose lack
means (though with a substantial amount of of sexual desire had persisted. Fourteen of these
variance). women had gone through natural menopause. They
were randomly assigned to either estrogen implant
Premenopausal women with endocrine abnormalities (40 mg) or estrogen-plus-testosterone implant (estro-
Miller et al. (2001) reported on the abnormally low gen 40 mg and testosterone 50 mg). The mean peak
androgen levels in women with hypopituitarism. In a serum total testosterone levels following implant

104
Chapter 5: The behavioral correlates of testosterone

slightly exceeded the upper limit of the normal range administration in the primary outcome measure, the
(i.e. 3 nmol/l). On average, the combined implant number of satisfying sexual episodes, and in a number
group showed improvement in sexual desire and of other measures including sexual desire. In contrast
sexual enjoyment within the first six weeks. The to earlier studies using im injection of testosterone,
estrogen only group did not report improvement, the use of transdermal testosterone administration
but when subsequently given an additional testoster- produced plasma levels of free testosterone and bioac-
one implant, they showed the same improvement as tive testosterone that remained within the physio-
the original combined group. No information was logical range for women. Modest but significant
given about the effects on mood or energy. correlations were found between increase in free tes-
Myers et al. (1990) studied 40 naturally postme- tosterone and improvements in several aspects of
nopausal women who were randomly assigned to four sexual function.
groups of 10 subjects each: P (Premarin only), PP
(Premarin plus Provera), PT (Premarin plus methyl- Surgical menopause
testosterone), and PL (placebo), and assessed over a Two studies will be considered closely (for a more
10-week period with both self-ratings and laboratory comprehensive review of the earlier literature, see
measurement of VPA response to erotic stimuli. Bancroft 2003). In the first study, Sherwin et al.
There were no group differences in treatment effects (1985) investigated women who were about to
on any of the sexuality variables, including VPA, undergo hysterectomy and bilateral ovariectomy.
except for masturbation. This showed a trend towards A one-month baseline assessment preceded the sur-
higher frequency and a significant increase in enjoy- gery: an unusual feature in the now extensive litera-
ment of masturbation in the PT group. There was a ture on surgical menopause. Post-operatively, women
trend (p = 0.06) towards group differences in mood were assigned randomly to one of four treatment
but, unfortunately, no further details were given of groups: estrogen only (E), testosterone only (T),
this measure. estrogen plus testosterone (E+T), or placebo. These
Davis et al. (1995) studied 34 postmenopausal were given in monthly injections for three months.
women, all but 2 of whom had gone through a natural All subjects then received one month of placebo,
menopause. All had shown intolerance of or inad- following which they were crossed over to one of the
equate response to oral HRT. They were randomly other three treatment groups. Women reported sig-
assigned to either estradiol implant or estradiol plus nificantly higher levels of sexual desire, sexual fanta-
testosterone implants, administered every three sies and sexual arousal during the E+T and
months for two years. Women with specific com- T conditions than during either the E or placebo
plaints of low sexual desire were excluded because it conditions. They did not differ in measures of sexual
was considered unethical to randomly assign them to activity with partner or orgasm. This is the only study
estradiol only! Women in both groups showed sig- in which testosterone administration on its own has
nificant improvement in sexuality measures, and, for been evaluated in ovariectomized women. It is also
most of the variables, the estradiol plus testosterone noteworthy because it focused on the immediate post-
group improved significantly more than the estradiol operative period in women who were not reporting
group, except that towards the end of the two-year significant sexual or mood problems pre-operatively.
study period there was a decline in their measures of Shifren et al. (2000) studied women who had
sexuality. This was attributed to a reduced frequency undergone hysterectomy and bilateral ovariectomy
of implants because of continuing supraphysiological from 1 to 10 years previously. In contrast to the
levels of testosterone. This suggests that some degree previous study, all had impaired sexual function and
of desensitization to the testosterone had occurred. all had been on Premarin, at least 0.625 mg daily for
More recently, Shifren et al. (2006) reported a at least two months when recruited for the study. All
large placebo-controlled trial of testosterone patches subjects continued on the same dose of oral estrogen
(300 mg) in naturally postmenopausal women meet- through the study. After a 4-week baseline assess-
ing the DSM-IV criteria for hypoactive sexual desire ment, they were given daily transdermal patches with
disorder; 273 women received placebo and 276 testos- placebo, 150 mg, or 300 mg of testosterone as the daily
terone. The average age was 54 years. Women showed dose, each for 12 weeks, with the order of presenta-
significant improvement from testosterone tion randomized. Sexuality was assessed using the

105
Chapter 5: The behavioral correlates of testosterone

Brief Index of Sexual Functioning for Women (BISF- Until recently, little research attention has been
W; Taylor et al. 1994). In spite of a substantial placebo paid to identifying different patterns of sexual desire
response, there was significantly more improvement and response in women. Of particular relevance is the
with the higher testosterone dose than with placebo question of what it is that is desired, which was
on measures of frequency of sexual activity, and pleas- considered earlier (in Section 5.4.1). Variations in
ure/orgasm, though not for sexual desire or arousal the importance of these patterns may account for
(the opposite pattern to that reported by Sherwin much of the variability in womens sexuality. It is also
et al. 1985). Mood was significantly improved with conceivable that certain patterns of sexual desire (e.g.
the higher testosterone dose for ratings of both desire for pleasure) may be more influenced by tes-
depression and positive well-being. The transder- tosterone than others (e.g. desire to be desired). (See
mal route used in this study has the advantage of Bancroft and Graham 2011 for further discussion of
producing more physiological and more stable serum this issue.)
testosterone levels than the im routes used in most
earlier studies, where supraphysiological peaks soon
after injection are followed by gradual decline. 5.4.2 Testosterone, mood and
Although the design of these two studies was
crucially different in a number of ways, there are aggression in women
some interesting contrasts in the findings. A more 5.4.2.1 Mood
substantial placebo effect was reported by Shifren In premenopausal women there is the impact of the
et al. (2000) than by Sherwin et al. (1985), and this menstrual cycle, and many women experience a cycle-
was more marked in the younger women. Thus, Shif- related variation in mood, which in its more prob-
ren et al. found no difference between placebo and lematic form is referred to as premenstrual syn-
active treatment on any variable in women younger drome or PMS. There is limited and inconsistent
than 48 years; the overall significant effects depended evidence that the cyclical variation in testosterone,
on the older women. The explanation for this age which tends to be at its highest during the middle
effect is not clear. It may reflect that for younger third of the cycle, may be associated with this cyclical
women loss of sexual interest or enjoyment is more mood pattern. In a small study of 11 women with
problematic and hence the expectation of, or need for severe premenstrual irritability and dysphoria (PMS)
improvement, greater. In the study by Sherwin et al., and 11 age-matched controls with no premenstrual
the less striking placebo effect may reflect the fact that problems, free testosterone levels were significantly
women were recruited before they had established any higher in the PMS group in the follicular phase,
post-operative decline in sexual interest. In contrast, around ovulation and in the luteal phase. Dehydro-
such decline was an inclusion criterion for the Shifren epiandrosterone levels were higher in the PMS group
et al. study. This emphasizes the importance of pla- around ovulation (Erikkson et al. 1992). In contrast,
cebo control in studies where subjects are seeking a Bloch et al. (1998) compared 10 women with PMS
therapeutic effect. and 10 controls. The women with PMS had signifi-
Since the report by Shifren et al. (2000), there have cantly lower total and free testosterone plasma levels
been four further studies of similar size, one involving with a blunting of the normal periovulatory peak.
European and Australian women (Davis et al. 2006), Premenstrual syndrome is a complex condition,
and three in the USA (Braunstein et al. 2005; Buster resulting from a variety of mechanisms which vary
et al. 2005; Simon et al. 2005). These are considered in importance from one woman to another (Bancroft
further in Chapter 23. Here again, in all these studies 1993). We should, therefore, not be surprised by
the results are presented only as mean change per inconsistencies of this kind, although it might be
treatment group, thus obscuring the proportion of informative to identify the relevant differences
women who were successfully treated. It is of crucial between the two samples.
importance to the field, from both a theoretical and In women the perimenopause is a time of
clinical perspective, to identify those women who increased vulnerability to depression. In the large
respond to testosterone therapy, and establish in what longitudinal Study of Womens Health Across the
ways they differ from those women who do not Nation (or SWAN), 3302 women entered the study
respond. aged 42 to 52, when premenopausal or early

106
Chapter 5: The behavioral correlates of testosterone

perimenopausal (Bromberger et al. 2010). Assays of of well-being. In a more recent national survey of
most reproductive steroids, including testosterone, women in heterosexual relationships, one of the best
and assessment of depression, using the CES-D (Radl- predictors of sexual distress was a marker of general
off 1977), were carried out annually for the next eight emotional well-being (Bancroft et al. 2003). This
years. A logistic regression, accounting for most of underlines the importance of mood to sexuality, and
the key variables assessed, showed that higher testos- the importance of sexuality to mood, particularly in
terone levels were significantly associated with higher women. Although it is well known in the psychiatric
odds of a CES-D score over 16. However, the signifi- field that sexual interest tends to be low in depressive
cance of this testosterone effect was less than that illness, this relationship in a more general sense has
found for menopausal status, ethnicity, age, education only recently started to receive attention (e.g. Davison
level, social support and occurrence of upsetting life et al. 2009).
events. Two small studies of women diagnosed with The next source of evidence comes from iatro-
major depression have shown increased testosterone genic lowering of testosterone in women. Oral con-
compared with controls. Baischer et al. (1995) traceptive use is commonly associated with negative
assessed 20 women (mean age 32.5) diagnosed with mood change, and this change, together with the
major depressive disorder, and compared them with negative sexual effects considered earlier, has been
an age-matched control group. They were assessed shown to be the most important predictor of discon-
twice; before and during treatment with clomipra- tinuation of oral contraceptives (Sanders et al. 2001).
mine. Before treatment, plasma testosterone was sig- Oral contraceptives also predictably lower testoster-
nificantly higher in the depressed women than the one levels, though to a variable extent. However, in
controls; after treatment testosterone levels were still the study of women starting on oral contraceptives,
higher than controls but the difference was no longer considered earlier, in which some association between
significant. Weber et al. (2000) reported on 11 reduced testosterone levels and sexual interest was
severely depressed women (aged 2877, mean 48.1) observed, there was no correlation between reduction
who were compared with 11 age-matched controls. in testosterone and negative mood change (Graham
Testosterone, androstenedione and DHT, together et al. 2007).
with cortisol, were all significantly higher in the Bilateral ovariectomy, as considered earlier, pro-
depressed women. The authors concluded that their duces substantial reductions in plasma testosterone.
findings were best explained as a consequence of But there is no consistent evidence of the effect that
over-stimulation of the adrenal glands. Other studies this has on well-being or mood; the surgical proced-
were briefly reviewed by Bromberger et al. (2010), ure is likely to produce improvements in well-being in
and they concluded that overall there has been con- so far as it resolves the pre-operative health problems.
siderable inconsistency on this issue. In the study reported by Aziz et al. (2005) in which
Let us look briefly at the relation between andro- 106 women had ovariectomy and hysterectomy
gens and mood in those studies we have already followed by estrogen replacement therapy, well-being
reviewed in relation to sexuality. In the large Austra- was assessed pre-operatively and at one year post-
lian survey (Davis et al. 2005), the relevant findings operatively. There was no correlation between post-
were published in a separate paper (Bell et al. 2006). operative levels of androgens and well-being. Beller-
This reported, in the younger age group, a significant ose and Binik (1993) compared five groups of women
association between DHEAS and vitality. So far, this aged 3555 years: three groups of ovariectomized
group has not indicated whether there was any asso- women, one group with no hormone replacement,
ciation between vitality and sexual function or one with estrogen only and one with estrogen plus
sexual well-being in this sample. Cawood and Ban- testosterone; the fourth group had hysterectomy only;
croft (1996), in a comparatively small study of women and the fifth was a control group of women with no
aged 4060 years, found that the only hormone sig- surgery. Although there were differences in sexual
nificantly correlated to well-being was DHEA. No interest and response, there were no differences in
significant associations between DHEA, DHEAS, mood across the five groups.
androstenedione or testosterone and measures of Sherwins prospective study of women undergoing
sexuality were found; however, one of the best pre- hysterectomy and ovariectomy, considered earlier (in
dictors of quality of sexual life was the womans state Section 5.4.1.4), gives us more controlled evidence of

107
Chapter 5: The behavioral correlates of testosterone

hormonal effects. This showed that testosterone, but possible that plasma testosterone levels might reflect
not estrogen administration, countered negative other aspects of the womans life, such as increased
effects of the surgery on sexual desire (Sherwin et al. stress. But this should be more apparent in levels of
1985). The effects of testosterone and estradiol on adrenal androgens, such as androstenedione or
mood and energy were reported in two additional DHEA. In the SWAN study these were not significant
papers. Sherwin and Gelfand (1985a) showed that predictors of depression. Hopefully, further research
mood was significantly better with all three hormone will resolve this issue.
regimes (T, E+T, and E) compared to placebo. The
testosterone-only group also had significantly higher 5.4.2.2 Aggression and assertiveness
hostility scores than the other three groups. In the Not surprisingly, the literature on testosterone and
third paper (Sherwin and Gelfand 1985b), it was aggression in women is much more limited than in
reported that energy level, well-being and appetite men. Only one of the studies involving testosterone
were significantly higher in the two groups receiving administration to women, considered earlier, reported
testosterone than in the estrogen-only or placebo any effect relevant to aggression. Sherwin and Gelfand
groups. Here again, the relations between these mood (1985a) found that ovariectomized women who
and energy effects and sexual effects have not been received testosterone only, rather than testosterone
reported, and we cannot therefore judge the extent to plus estrogen, estrogen or placebo, had significantly
which sexual improvement may have been secondary higher hostility scores than the other three groups.
to mood improvement or vice versa. Two studies assessed women who had shown vio-
Shifren et al. (2000) in their study, considered lent behavior. Ehlers et al. (1980) compared young
earlier (in Section 5.4.1.4), of women presenting with female clinic attendees with a history of violence with
impaired sexual function after bilateral ovariectomy a non-violent control group attending the same clinic.
and hysterectomy, found that in addition to improve- Those with a history of violence had significantly
ments in sexual function, there were significant higher plasma testosterone levels. Dabbs et al. (1988)
improvements in both depression and positive well- compared inmates in a womens prison with convic-
being with the higher testosterone dose. Goldstat tions for different types of crime. They divided them
et al. (2003), in assessing the effects of testosterone into five groups: Unprovoked Violence (UV) (n = 15),
administration for the treatment of low sexual desire, Defensive Violence (DV) (n = 5), Theft (n = 40),
found significant improvements in general well-being Drugs (n = 14) and Other (n = 10). Those convicted
as well as sexuality. of Unprovoked Violence had the highest mean saliv-
A number of placebo-controlled studies have ary testosterone (2.63 ng/100 ml) whereas the Defen-
evaluated the effects of DHEA administration on sive Violence group had the lowest (1.48 ng/100 ml).
mood; the earlier studies were reviewed by Buvat This was significantly higher in the UV group than
(2003). The most consistent benefit observed has been the DV group and also the Theft group (1.94 ng/100
improved energy and well-being, with less consistent ml). Apart from the UV group, the other four groups
effects on sexuality. Schmidt et al. (2005) reported a did not differ significantly from each other.
double-blind, placebo-controlled, crossover study of Gladue (1991) measured testosterone, free testos-
DHEA (for six weeks) in the treatment of 23 women terone and estradiol in samples of male and female
and 23 men presenting with midlife onset of depres- students, and aggressive tendencies were assessed by
sion. In comparison to placebo, there was a significant a modification of the Olweus Multifaceted Aggression
improvement in mood and sexuality in both women Inventory (Olweus 1975), originally designed for use
and men. In a more recent review, Panjari and Davis with adolescents. He found significantly higher aggres-
(2010) concluded that there was no consistent benefit sion scores in the men than in the women, with women
from DHEA administration for either mood or sexu- being more likely to avoid confrontation. Testosterone
ality across the various studies. However, they con- and estradiol were positively correlated with several
fined their attention to postmenopausal women. indices of aggressive behavior in men, but were nega-
Apart from the SWAN study, none of these stud- tively correlated with those same indices in women.
ies gave any indication that testosterone might Harris et al. (1996) measured salivary testosterone
increase the likelihood of depression, and there is no in male and female students, and used a number of
obvious mechanism by which it would do so. It is questionnaires to assess two personality factors, an

108
Chapter 5: The behavioral correlates of testosterone

aggression factor and a pro-social factor. The men sexual partners was also correlated with testosterone
scored higher than the women on the aggression and estradiol. In contrast, high ranking by ones peers
factor and lower on the pro-social factor. In contrast was negatively correlated with androgens, particularly
to Gladues (1991) findings, the aggression factor was with androstenedione. Cashdan made an interesting
positively correlated, and the pro-social factor nega- interpretation, based on the fact that 62% of her
tively correlated, with testosterone in both men and sample were practicing Mormons. She distinguished
women. between women who sought success and achievement
Van der Pahlen et al. (2002) studied 33 healthy through their own actions, and were hence more
women, mean age 28.3, with regular cycles. Each competitive with both men and women, and women
woman had one blood sample taken mid-cycle at 6 who sought high parental investment from their
p.m. This was assayed for total and free testosterone, sexual partners, traditional family values and pre-
androstenedione, DHT and DHEA. Their aggressive marital chastity. Apparently, practicing Mormons at
propensities were assessed with the BussPerry Utah University are more likely to fit the second
Aggression Questionnaire (Buss and Perry 1992). pattern; hence their tendency to devalue the women
This has four subscales: physical aggression, verbal with higher androgen-related behaviors.
aggression, anger and hostility. A composite An intriguing situation in which to consider the
aggression score derived from the four subscales effects of testosterone on aggression in genetic
was compared to the hormone levels. Three hor- females is in the transition of female to male trans-
mones accounted for a significant proportion of the gendered individuals. Part of this process involves
variance: free testosterone 24%, DHT 16% and administration of testosterone at dosages that would
androstenedione 14%. Two of the subscales showed typically be used for treating hypogonadal males. Van
significant correlations with hormones: verbal Goozen et al. (1995) reported a study in which 35
aggression with free testosterone (r = 0.49; p female-to-male and 15 male-to-female transgendered
< 0.004), and anger with DHT (r = 0.38, p = 0.03). individuals were assessed shortly before and three
The authors concluded that other androgens in add- months after the start of cross-gender hormonal treat-
ition to testosterone were relevant to aggression. ment. The female-to-male individuals showed an
Cashdan (2003) studied women students, with regu- increase in aggression proneness, sexual arousability
lar menstrual cycles and not using hormonal contracep- and spatial ability. The male-to-female individuals,
tion, who were asked to complete a form on a total of 10 who received an antiandrogen (cyproterone acetate)
occasions on days when they felt that they had behaved and estrogen, showed reduced aggression proneness
in a competitive manner. For this purpose competi- and sexual arousability and improved verbal ability.
tion was defined very broadly: e.g. trying to improve Overall, however, self-ratings of mood and well-being
ones position relative to someone elses. On the form showed little negative change, perhaps reflecting their
they were asked to describe how they had felt and welcome for these crucial hormonal treatments in the
behaved. Blood samples were taken on one day early transgender process.
in the follicular phase, and were assayed for total and In summary, in women, we have little evidence
free testosterone, estradiol, androstenedione and corti- that testosterone and estradiol have direct effects on
sol. Women with low levels of androstenedione and mood, and somewhat inconsistent evidence that tes-
total testosterone were less likely to express their com- tosterone can enhance aggression.
petitive feelings overtly, while women with high levels of
androstenedione were more likely to show their com-
petitiveness with verbal aggression. 5.4.3 Summary of evidence in the female
In an earlier study, involving women students There are good reasons for thinking that testosterone
from the same university (Utah), Cashdan (1995) has a role in the sexuality of women, but the evidence
compared womens ranking of themselves within the is inconsistent and confusing. There are a number of
student group, with how they were ranked by other possible explanations for this:
students participating in the study. She found that 1. Women may vary in the extent to which their
those women who over-ranked themselves in com- sexuality is influenced by testosterone. There is
parison to how they were ranked by others had higher much indirect evidence to support this: the
testosterone and estradiol levels. The number of possibility that oral contraceptives reduce sexual

109
Chapter 5: The behavioral correlates of testosterone

interest by lowering free testosterone but only in a relationship between testosterone and sexuality
proportion of women; the negative sexual effects may be more apparent in women who are not
that antiandrogens have in a proportion of experiencing significant relationship problems.
women; the finding that sexual interest is lowered
in many but not all women after ovariectomy 5.5 Comparison of testosterone
(approximately 50%). Sherwin (1988), who has
conducted some of the most important research in effects in men and women
this area, commented on the considerable inter- What are the principal points that emerge from a
subject variability in ovariectomized womens comparison of testosterone effects in men and
responses to testosterone in her studies. It is women?
striking that, in the extensive literature on 1. The evidence for an activating effect of
hormone replacement in women, virtually no testosterone on sexual interest and response is
attention has been paid to individual variability. more consistent for men than for women. It has
Without any attempt to control for such been proposed that this reflects greater individual
variability we should expect the kind of variability of behavioral responsiveness to
inconsistency across studies that we have androgens among women. There obviously is
encountered, particularly when sample sizes are variability among men but it appears to be of a
small. Given that testosterone effects in the male much smaller magnitude.
are essential for reproduction, we should expect 2. Those women who are behaviorally responsive to
less variability in men. In women, however, such testosterone respond to levels of testosterone that
testosterone effects are probably irrelevant to would be totally ineffective in men. This greater
reproduction, and hence genetic variability would sensitivity to testosterone in women is apparent in
not be surprising. behavioral and other CNS responses. It is less clear
The variability in the association between testosterone whether it is as apparent in the anabolic and skin
and mood and aggression in women is not effects of testosterone. The limited evidence we
fundamentally different from that found in men. have about the distribution of testosterone
Hence we need to consider whether there are basic receptors or ARs in the brain only adds to this
differences in the testosterone-dependent puzzle. It so far indicates many more ARs in the
mechanisms involved in sexual response and mood/ male than the female brain, with some brain areas
aggression responses. where the gender difference is particularly marked
2. We remain uncertain about the extent to which (see Section 5.2.2). Why do men, who need so
observed behavioral effects of exogenous much more testosterone for their sexually-relevant
testosterone are direct androgen effects or indirect brain effects, have so many more testosterone
effects resulting from increased availability of receptors? The distribution of ARs, while overall
bioactive estrogen, resulting both from the at a much lower level, is more variable among
conversion of testosterone to estradiol, and women. This in itself is consistent with the idea of
reduced binding of estradiol in the presence of variable testosterone responsiveness in women.
increased testosterone. But this does not resolve the basic mystery.
3. The sexuality of women is powerfully influenced 3. Men and women may differ in the relationship
by mood, energy and well-being, and these aspects between affect and sexuality. Although studies of
are affected by a wide variety of factors. Although androgen withdrawal and replacement in men
possible effects of testosterone on mood, as well as show that mood, energy and well-being are
sexuality, have been assessed in many of the affected, the impact on these variables appears to
studies considered, very few have directly be more influential and predictable in women
considered the extent to which mood influences than in men. The reasons for this are poorly
sexuality. In addition to depression, the sexuality understood. In general, the relationship between
of women is powerfully influenced by other mood and sexuality has received very little
psychological mechanisms. As in an early study of attention until recently, and we still have a lot to
oral contraceptive users (Bancroft et al. 1980), the learn.

110
Chapter 5: The behavioral correlates of testosterone

4. A fundamental difference between male and the most extreme example of abnormality in the AR
female mechanisms of androgen production may gene, although it may involve a variety of mutations
be relevant. More than 90% of testosterone in the (see Chapter 3). Interviews with 15 women with CAIS
male is produced by the testes. By contrast, a were reported recently (Bancroft 2009), along with a
substantial proportion of androgen production in review of other studies of women with CAIS; Money
women is from the adrenal glands, and hence and Ehrhardt (1972; 10 women), Vague (1983;
increased adrenal androgens can be expected in 7 women), Wisniewski et al. (2000; 14 women) and,
states associated with increased adrenal activity, the largest study, Minto et al. (2003; 59 women).
such as anxiety, stress or depression. No evidence Across these studies, apart from an increased likeli-
of increased testosterone in negative mood states hood of experiencing difficulties with a small or tight
in men has been found. vagina early in their first sexual relationship,
5. In the male, the evidence supports the idea of a these women present a varied picture of female sexu-
threshold above which increased testosterone ality, not clearly different from normal women,
levels have little additional effect relevant to although shifted towards the less responsive end of
sexuality, and below which signs of androgen the range. Many of them experienced orgasm, often
deficiency are likely to occur. It is possible that a without difficulty. Thus it would seem that orgasm is
small minority of men are sensitive to higher not testosterone dependent. Unfortunately, this
levels in terms of manic or aggressive response, evidence does not give us any clear guidance towards
but this does not seem to apply to male sexuality. testosterone-dependent aspects of womens sexuality.
In women we are faced with a dilemma. As yet, What other effects do polymorphisms of the AR
the only studies showing a significant correlation gene have in men and women? The AR gene is located
between plasma testosterone and sexual interest on chromosome Xq1112, a polymorphic polygluta-
have involved women using oral contraceptives, mate in the AR encoded by the nucleotides cysteine,
whose testosterone levels are substantially adenine and guanine; hence CAG (Westberg et al.
lowered. If this has general validity, it would 2001; see Chapter 3).
suggest that most women have much more In the male, abnormalities range from severe con-
testosterone than they need. It is already ditions, such as X-linked spinobulbar muscular atro-
remarkable that testosterone can have effects in phy, to varying degrees of hypogonadism, and in
women at levels that are no more than a 10th of some cases impaired spermatogenesis (Zitzmann
male levels: a gender difference that requires and Nieschlag 2003). However, in a recent Brazilian
explanation. If in fact women need only a small study (Andersen et al. 2010), CAG AR repeats were
proportion of that 10% then it is even more genotyped in 79 men with erectile dysfunction (ED)
remarkable. Set against this is the evidence that and 340 controls. There was no association between
supraphysiological levels of testosterone in the AR CAG repeat polymorphism and ED. An
women, that often result from im injection of important point that is emerging is that rather than
testosterone esters, are associated with activational consider the effects of plasma testosterone levels per
effects; though it is not clear whether such effects se, we should be looking at the interaction between
are specifically sexual or more non-specifically plasma testosterone and the length of the CAG repeat
arousing, or a mixture of both. Furthermore, such on the AR gene. As an example, the effects of testos-
effects are typically of limited duration, with the terone replacement on prostate growth in hypogona-
need for increasing frequency of testosterone dal men are potentially problematic in men with the
injections to maintain them. This suggests the short CAG repeats, and hence lower doses of testos-
development of tolerance, and points to a terone would be advised in such cases.
desensitization effect. Various behavioral patterns, which are clearly dif-
ferent in males and females, have nevertheless shown
How does our increasing knowledge of the andro- inconsistent correlations with plasma testosterone
gen and estrogen receptors help in our attempts to levels, which may reflect differences in CAG length.
understand the role of testosterone in men and women? In an interesting study of 301 adolescent males (mean
The starting point is the condition of complete age 14.4 years) in which non-aggressive and aggres-
androgen insensitivity syndrome (CAIS), possibly sive risk taking, dominance, depression, and self-

111
Chapter 5: The behavioral correlates of testosterone

esteem were among the variables assessed, Ver- were associated with higher testosterone levels.
meersch et al. (2010) found that free testosterone Whereas in men the inhibitory effect is mediated via
was more strongly related to both non-aggressive the hypothalamus and LH secretion, in women the
and aggressive risk taking in boys with shorter CAG stimulation is due to a direct effect of ARs in the
repeat lengths, and was only related to dominance in adrenal gland and ovaries. For some reason, which
boys with short CAG lengths. They also found that Westberg et al. could not explain, this stimulatory
free testosterone was negatively associated with effect was not relevant to DHEAS (or presumably
depressive symptoms and self-esteem in boys with DHEA which they did not assay), and this is the
longer CAG lengths. Simmons and Roney (2011) principal androgen secreted by the adrenal glands.
considered the contrast between mating effort and The mystery of DHEA is further deepened! The poly-
survival in men and commented that an expanding morphisms of the two estrogen receptors, ERa and
body of research suggests that circulating androgens ERb were also explored. Short CAG repeats of the
regulate the allocation of energy between mating and ERb were associated with high levels of serum testos-
survival effort in human males, with higher androgen terone and free testosterone. Westberg et al. suggested
levels promoting greater investment in mating effort. The possibility that a relatively pronounced influ-
They studied 138 undergraduate students, measuring ence of androgens during development leads to an
upper body strength, BMI, measures of dominance (a increase in serum androgen levels in adult women
subscale of the Self-Perceived Social Status Scale; should be considered. As yet, however, gene poly-
N. R. Buttermore and L. A. Kirkpatrick (unpublished) morphisms have not helped us to explain any of the
Distinguishing dominance and prestige: two distinct major variations in female sexuality.
pathways to status) and the Sociosexual Orientation
Inventory (SOI; Simpson and Gangestad 1991), which 5.5.1 Some hypothetical ideas about
gives a measure of propensity for casual sex. Subjects
with shorter CAG repeat lengths had greater upper testosterone and gender differences
body strength and scored higher on measures of Two possible explanatory models of the role of tes-
dominance and prestige. Plasma testosterone levels tosterone in women can be considered. The first is
were not predictive of any of these measures, nor of that women vary in their sensitivity to testosterone,
SOI. The authors suggested that long-term testoster- and for most women a high sensitivity is associated
one exposure interacting with the AR gene poly- with a very low threshold level, sufficient to main-
morphisms would account for more of the variance tain the testosterone effects. In addition there could
in these measures than current testosterone levels, be women who have much lower sensitivity to
and concluded that such effects related to long-term testosterone, possibly as a result of some degree of
mating effort rather than casual sex. desensitization in early development along male
Thus we are starting to understand and hopefully lines. They will experience a decline in testosterone-
find a solution for much of the variability in the dependent responses when the testosterone level is
literature on testosterone and male sexuality and lowered, as by oral contraceptives or ovariectomy. In
mood. However, as we have already encountered, contrast, the high-sensitivity women will still have
the variability in the relation between testosterone, enough testosterone after such iatrogenic reductions.
sexuality and mood is substantially greater in women. The second model is that some women are
To what extent will assessment of AR polymorphisms responsive to testosterone effects on their sexuality,
help us grapple with this? whereas others are not. That would assume that for
Interestingly, there are important gender differ- many women, possibly the majority, sexuality is not
ences in this respect. Westberg et al. (2001) carried testosterone dependent, whereas for some there is a
out genotyping and hormone assays in 270 women testosterone-dependent component added. This
from a large population-based Swedish cohort. They requires consideration of how the sexuality of the
observed associations between serum androgen levels non-testosterone-dependent woman differs from that
and CAG repeats, but with some striking differences of the testosterone-dependent woman.
from those observed in men. The major influence of In the next section I will propose two hypotheses
ARs on testosterone production in women is stimula- which have the potential to be tested and may lead to
tory, rather than inhibitory as in men. Short repeats a better understanding of the complex role of

112
Chapter 5: The behavioral correlates of testosterone

testosterone. In the second hypothesis that focuses on supraphysiological levels of testosterone was reported
women, I will be using the second of the two explana- in several of the HRT studies in women reviewed
tory models I have just described. earlier. This suggests that such desensitization might
occur later in life also, at least to some extent. If there
5.5.1.1 The Desensitization hypothesis is any validity in this desensitization hypothesis, it is
Men have far more testosterone than they need for important that we know about it, as it could be highly
activational effects in the brain. On the other hand, relevant to long-term effects of sustained supraphy-
they need high levels to establish and maintain the siological levels of testosterone in older women. How-
masculinizing effects in the rest of the body, including ever, we should also keep in mind the possibility that
spermatogenesis. How much they need for maintain- there may be a decline in testosterone receptor sensi-
ing peripheral sexual responses, such as erection, is tivity in women as they age, comparable to that found
not clear. But it could possibly be higher than needed in men.
for the brain effects. It is therefore postulated that, at I first wrote about this desensitization hypothesis
some stage of development, a desensitization of ARs in 2002 (Bancroft 2002), and have written about it
in the brain occurs. This allows high levels of several times since. However, I have not yet encount-
testosterone to be maintained in the circulation with- ered any reaction to it in the literature, either positive
out excessive and maladaptive activation of the testos- or negative. I am aware that the hypothesis is crude, as
terone-dependent system in the brain. As this is I am unable to provide any plausible explanation of
necessary for normal male sexual function and hence how such desensitization might occur. But I must leave
reproduction, which can be called the basic pattern that to those who have a better understanding of recep-
of male sexuality, we would not expect much genetic tor physiology. This takes us to the second hypothesis.
variation in the determinants of this desensitization
process. 5.5.1.2 The By-product hypothesis
Exposure to substantially higher levels of testoster- This assumes that there is a basic pattern of
one during fetal development and also during the first womens sexuality which, as with the basic pattern
few weeks postnatally could be responsible for desen- in the male, is necessary for reproduction, and hence
sitizing the CNS to testosterone effects in the male. is of fundamental importance. This may depend on
Although, as we have already considered, there are the womans motivation to be sexually desired by her
crucial differences in the control of androgens in partner and to accept his wish for vaginal intercourse.
women, it may be relevant to consider conditions in It may also involve the desire for the emotional intim-
which females are exposed to high androgen levels acy that is typically associated with penile-vaginal
during fetal development. The best example is con- intercourse. Awareness of its fundamental reproduct-
genital adrenal hyperplasia (CAH), particularly the ive purpose may also be a contributory factor. This
salt-losing variety that is associated with higher levels basic pattern is helped by the womans relatively
of testosterone during fetal development. This is not automatic vaginal lubrication response and special-
only associated with some degree of masculinization ized pain reduction mechanisms that result in vaginal
of behavior, but also low levels of sexual interest and penetration being painless. The vaginal response is
activity and low fertility (Meyer-Bahlburg et al. 2003). dependent on estradiol, although the determinants
Although in such cases there are a number of factors of the womans desire for this type of interaction are
which could impair normal sexual development, this not yet understood.
evidence is consistent with there being some degree of Thus I am proposing that there are two basic
desensitization to the high fetal levels of testosterone, patterns essential for reproduction; the male pattern
which fall and remain in the normal female range is testosterone dependent and involves insertion of
once the CAH is treated. the erect penis into the female partner, rewarded by
An interesting question is whether this hypothet- the associated pleasure, in particular with the orgasm
ical desensitization mechanism is an organizing that accompanies seminal emission. I would assume
effect of high testosterone levels, which is only opera- that both basic patterns are sufficiently fundamental
tive during early development, or whether such sup- to reproduction that there will be little genetic vari-
pression is possible if exposure to high levels occurs ation in their determinants. In addition, I am propos-
later in development. Evidence of tolerance to ing that, as examples of by-products using the

113
Chapter 5: The behavioral correlates of testosterone

concept introduced by Symons (1979), components


of the male pattern are evident in some women, and sexual arousal in men, and also contributes
to normal erectile function, spermatogenesis and
presumably reflecting variable genetic determination.
the capacity for seminal emission. Decline in
This concept has been persuasively argued by Lloyd
testosterone with age can contribute to the
(2005), amid much controversy, to explain the occur- complex effects of aging on male sexual function.
rence of orgasm in women. I am proposing that  Variability in the relationship between plasma
testosterone-dependent effects, mainly in the motiv- testosterone and male sexual function is
ation for sexual pleasure, are other by-products. It is attributable in part to the length of CAG repeats
also feasible that there are aspects of male sexuality in the AR gene. The effects of specific
that are by-products of the female basic pattern. testosterone levels in the male need to be
However, finding aspects of female sexual experi- assessed in interaction with CAG repeat length.
ence that can be shown to be dependent on testoster-  Whereas there is little evidence of a correlation
one is not going to be easy. between plasma testosterone levels and mood or
aggression in men, effects of supraphysiological
levels of testosterone indicate that a small minority
5.5.1.3 One way forward of men may be sensitive to the arousing effects of
While by no means simple, this theoretical model, testosterone, either as manic mood or aggression.
based on two principal hypotheses, is open to testing This presumably reflects a genetic variability not
in observational and clinical studies. It is possible that relevant to the effects of physiological
testosterone levels on male sexuality.
receptor polymorphisms, which are being intensively
 As with men, there is no consistent evidence of a
studied, will be of relevance. At this point it is not correlation between plasma testosterone levels
clear how we would use the recently acquired know- and sexual interest or response in women. The
ledge in this respect. effects of iatrogenic lowering of testosterone, e.g.
One way forward, at this point in time, is to focus by use of oral contraceptives, or bilateral
first on developing a typology of womens sexuality, ovariectomy, indicate that a proportion of women,
which aims to characterize at least two different types, but not all, experience a decline in sexual interest
which would then allow groups of women, who fit the and responsiveness. This has raised the possibility
characteristics of each type, to be compared on vari- that women vary markedly in their sensitivity to
ables that relate to testosterone effects, which would testosterone, or that some are responsive to
include AR gene polymorphisms, levels of plasma testosterone and others are not. Administration of
testosterone to women with lowered sexual
testosterone and other androgens, and other possible
interest often produces improvement.
indicators of androgen effects. This approach to a
 The majority of the relevant literature on women
typology of womens sexuality has recently been con- ignores the possibility of individual variability in
sidered at more length(Bancroft and Graham 2011). testosterone sensitivity and presents results as
group means. There has been no attempt to
5.6 Key messages identify potential markers of testosterone
 We have little understanding of how and where responsiveness in women.
testosterone works within the brain, either in  The main difference between men and women in
organization of brain development or as relation to testosterone is that plasma levels of
activational effects. There is limited evidence of testosterone in women are around 10% of those
the distribution of ARs and ERs in the human in the male, and testosterone administration can
brain. Evidence from non-human primates produce behavioral effects in women with
suggests certain areas in the subcortical brain dosages that would be totally ineffective in men.
where testosterone works principally by being The basis for this gender difference in sensitivity
aromatized to estradiol and other areas where it to testosterone has not yet been explained.
acts directly via ARs. There is also limited  The sources of testosterone and other androgens
evidence of ARs in the brain being less frequent and the control of their synthesis are clearly
and more variable across women than in men. different in men and women. Ninety percent of
 There is reasonably consistent evidence that testosterone in the male comes from the testes.
testosterone is necessary for normal sexual desire In the female around 25% comes from the ovary

114
Chapter 5: The behavioral correlates of testosterone

and 25% from the adrenal glands, with the Whereas in men the short gene inhibits
remainder derived from peripheral conversion of testosterone synthesis, in women it
androstenenedione or DHEA. Control of stimulates it.
testosterone production in the male is via the  Hypotheses have been proposed in this
hypothalamus and LH secretion. In the female it chapter to account for the major gender
is via ARs in the adrenal glands and ovaries. difference in testosterone levels and for the
 There is also a striking gender difference in the possible variability of testosterone
impact of the CAG repeat length of the AR gene. responsiveness in women.

5.7 References Anderson RA, Martin CW, Kung A,


Everington D, Pun TC, Tan KCB,
Bancroft J (1993) The premenstrual
syndrome a reappraisal of the
Abdelgadir SE, Roselli CE, Choate JVA, Bancroft J, Sundaram K, Moo- concept and the evidence. Psychol
Resko JA (1999) Androgen receptor Young AJ, Baird DT (1999) 7a- Med Suppl 24:147
messenger ribonucleic acid in brains Methyl-19-Nortestosterone
and pituitaries of male rhesus Bancroft J (1995) Are the effects of
(MENT) maintains sexual
monkeys: studies on distribution, androgens on male sexuality
behavior and mood in hypogonadal
hormonal control, and relationship noradrenergically mediated?
men. J Clin Endocr Metab
to luteinizing hormone secretion. Some consideration of the
84:35563562
Biol Reprod 60:12511256 human. Neurosci Biobehav Rev
Appelt H, Strauss B (1986) The 19:325330
Alder E, Cook A, Davidson D, West C,
psychoendocrinology of female Bancroft J (2002) Sexual effects of
Bancroft J (1986) Hormones, mood
sexuality: a research project. androgens in women: Some
and sexuality in lactating women.
German J Psychol 10:143156
Brit J Psychiat 148:7479 theoretical considerations. Fert
Aziz A, Bramstrom M, Bergquist C, Steril 77 (Suppl 4):S55S59
Alexander GM, Sherwin BB (1993) Sex
Silverstolpe G (2005) Bancroft J (2003) Androgens and
steroids, sexual behavior, and
Perimenopausal androgen decline sexual function in men and women.
selection attention for erotic stimuli
after oophorectomy does not In: Bremner W, Bagatell C (eds)
in women using oral contraceptives.
influence sexuality or Androgens in Health and Disease.
Psychoneuroendocrinology
psychological wellbeing. Fert Steril
18:91102 Humana Press, Totowa, NJ,
83:10211028 pp 259290
Almeida OP, Yeap BB, Hankey GJ,
Jamrozik K, Flicker L (2008) Low Bagatell CJ, Heiman JR, Rivier JE, Bancroft J (2009) Human Sexuality and
free testosterone concentration as a Bremner WJ (1994a) Effects of its Problems, 3rd edn. Churchill
potentially treatable cause of endogenous testosterone and Livingstone, Edinburgh
depressive symptoms in older men. estradiol on sexual behavior in
normal young men. J Clin Endocr Bancroft J, Cawood EHH (1996)
Arch Gen Psychiatry 65:283289 Androgens and the menopause: a
Metab 78:711716
Amiaz R, Seidman SN (2008) study of 40 to 60 year old women.
Testosterone and depression in Bagatell CJ, Heiman JR, Matsumoto Clin Endocrinol 45:577587
men. Curr Opin Endocrinol Diabet AM, Rivier JE, Bremner WJ (1994b)
Metabolic and behavioral effects of Bancroft J, Graham CA (2011) The
Obes 15:27828
high-dose exogenous testosterone in varied nature of womens sexuality:
Andersen ML, Guindalini C, Santos- healthy men. J Clin Endocr Metab unresolved issues and a theoretical
Silva R, Bittencourt LRA, Tufik S 79:561567 approach. Horm Behav 59:717729
(2010) Androgen receptor CAG
Baischer W, Koinig G, Hartmann B, Bancroft J, Sartorius N (1990) The
repeat polymorphism is not
Huber J, Langer G (1995) effects of oral contraceptives on
associated with erectile dysfunction
Hypothalamic-pituitary-gonadal well-being and sexuality. Oxford Rev
complaints, gonadal steroids and
axis in depressed premenopausal Reprod Biol 12:5792
sleep parameters: data from a
population-based survey. J Androl women: elevated blood testosterone Bancroft J, Davidson DW, Warner P,
32:524529 concentrations compared to normal Tyrer G (1980) Androgens and
controls. Psychoneuroendocrinology sexual behaviour in women using
Anderson RA, Bancroft J, Wu FCW 20:553559 oral contraceptives. Clin Endocrinol
(1992) The effects of exogenous
Bancroft J (1989) Human Sexuality and 12:327340
testosterone on sexuality and mood
of normal men. J Clin Endocr Metab its Problems, 2nd edn. Churchill Bancroft J, Sanders D, Davidson D,
75:15031507 Livingstone, Edinburgh Warner P (1983) Mood, sexuality,

115
Chapter 5: The behavioral correlates of testosterone

hormones and the menstrual cycle syndrome and controls. Biol transition. J Clin Endocr Metab
III. Sexuality and the role of Psychiat 43:897903 85:28322838
androgens. Psychosom Med Book AS, Starzyk KB, Quinsey VL Buss AH, Durkee A (1957) An
45:509516 (2001) The relationship between inventory for assessing different
Bancroft J, Sherwin B, Alexander GM, testosterone and aggression: a meta- kinds of hostility. J Consult Psychol
Davidson DW, Walker A (1991) analysis. Aggress Violent Behav 21:343349
Oral contraceptives, androgens, and 6:579599 Buss AH, Perry M (1992) The
the sexuality of young women. II. Boyd RA, Zegarac EA, Posvar EL, Flack Aggression Questionnaire.
The role of androgens. Arch Sex MR (2001) Minimal androgenic J Personal Soc Psychol 63:452459
Behav 20:121135 activity of a new oral contraceptive Buster JE, Kingsberg SA, Aguirre O,
Bancroft J, Loftus J, Long JS (2003) containing norethindrone acetate Brown C, Breaux JG, Buch A,
Distress about sex: a national survey and graduated doses of ethinyl Rodenberg CA, Wekselman K,
of women in heterosexual estradiol. Contraception 63:7176 Casson P (2005) Testosterone patch
relationships. Arch Sex Behav
Braunstein GD, Sundwall DA, Katz M, for low sexual desire in surgically
32:193208
Shifren JL, Buster JE, Simon JA, menopausal women: a randomized
Barrett-Connor E, von Mhlen DG, Bachman G, Aguirre OA, Lucas JD, trial. Obstet Gynecol 105:944952
Kritz-Silverstein D (1999) Rodenberg C, Buch A, Watts NB Buvat J (2003) Androgen therapy with
Bioavailable testosterone (2005) Safety and efficacy of a dehydroepiandrosterone. World
and depressed mood in older testosterone patch for the treatment J Urol 21:346355
men: the Rancho Bernardo of hypoactive sexual desire disorder
Study. J Clin Endocr Metab in surgically menopausal women. Caprara GV, Barbaranelli C, Comrey
84:573577 Arch Intern Med 165:15821589 AL (1992) A personological
approach to the study of aggression.
Beck AT, Ward CH, Mendelson M, Bromberger JT, Schott LL, Kravitz HM, Personality Individ Diff 13:7784
Mock J, Erbaugh J (1961) An Sowers M, Avis NE, Gold EB,
inventory for measuring depression. Randolph JF Jr, Matthews KA Carani C, Scuteri A, Marrama P,
Arch Gen Psychiatry 4:561571 (2010) Longitudinal change in Bancroft J (1990) The effects of
reproductive hormones and testosterone administration and
Behre HM, Bckers A, Schlingheider A,
depressive symptoms across the visual erotic stimuli on nocturnal
Nieschlag E (1994) Sustained
menopausal transition. Arch Gen penile tumescence in normal men.
suppression of serum LH, FSH and
Psychiatry 67:598607 Horm Behav 24:435441
testosterone and increase of high-
density lipoprotein cholesterol by Brown WA, Davis GH (1975) Serum Carani C, Granata ARM, Bancroft J,
daily injections of the GnRH testosterone and irritability in man. Marrama P (1995) The effects of
antagonist cetrorelix over 8 days in Psychosom Med 37:87 testosterone replacement on
normal men. Clin Endocrinol nocturnal penile tumescence and
40:241248 Buena F, Swerdloff RS, Steiner BS, rigidity and erectile response to
Lutchmansingh P, Peterson MA, visual erotic stimuli in hypogonadal
Bell RJ, Donath S, Davison SL, Davis Pandian MR, Galmarini M, Bhasin
SR (2006) Endogenous androgen men. Psychoneuroendocrinology
S (1993) Sexual function does not 20:743753
levels and well-being: differences change when serum testosterone
between premenopausal and levels are pharmacologically varied Carney A, Bancroft J, Mathews A
postmenopausal women. within the normal male range. Fertil (1978) Combination of hormonal
Menopause 13:6571 Steril 59:11181123 and psychological treatment for
Bellerose SB, Binik YM (1993) Body female sexual unresponsiveness: a
Burger H, Hailes J, Nelson J, Menelaus comparative study. Brit J Psychiat
image and sexuality in M (1987) Effect of combined
oophorectomized women. Arch Sex 132:339356
implants of oestradiol and
Behav 22:435460 testosterone on libido in Cashdan E (1995) Hormones, sex and
Blackburn IM, Loudon JB, Ashworth postmenopausal women. Brit Med J status in women. Horm Behav
CM (1977) A new scale for 294:936937 29:354366
measuring mania. Psychol Med Burger HG, Dudley EC, Cui J, Cashdan E (2003) Hormones and
7:453458 Dennerstein L, Hopper JL (2000) competitive aggression in women.
Bloch M, Schmidt PJ, Su T-P, Tobin A prospective longitudinal study of Aggress Behav 29:107115
MB, Rubinow DR (1998) Pituitary- serum testosterone, Cawood EHH, Bancroft J (1996)
adrenal hormones and testosterone dehydroepiandrosterone sulfate, Steroid hormones, the menopause,
across the menstrual cycle in and sex hormone-binding globulin sexuality and well-being of women.
women with premenstrual levels through the menopause Psychol Med 26:925936

116
Chapter 5: The behavioral correlates of testosterone

Christiansen K, Knussman R (1987) Davis SR, McCloud P, Strauss BJG, Ehlers CL, Rickler KC, Hovey JE (1980)
Androgen levels and components of Burger H (1995) Testosterone A possible relationship between
aggression in men. Horm Behav enhances estradiols effects on plasma testosterone and aggressive
21:170180 postmenopausal bone density and behavior in a female outpatient
Coenen CMH, Thomas CMG, Borm sexuality. Maturitas 21:227236 population. In: Girgis M, Kiloh LG
GF, Hollanders JMG, Rolland R Davis SR, Davison SL, Donath S, Bell RJ (eds) Limbic Epilepsy and Dyscontrol
(1996) Changes in androgens (2005) Circulating androgen levels Syndrome. North Holland
during treatment with four low- and self-reported sexual function in Biomedical Press, New York,
dose contraceptives. Contraception women. JAMA 294:9196 pp 183194
53:171176 Davis SR, van der Mooren MJ, van Erikkson E, Sundblad C, Lisj P,
Corts-Gallegos V, Castaneda G, Lunsen RH, Lopes P, Ribot C, Rees Modigh K, Andersch B (1992)
Alonso R, Sojo I, Carranco A, M, Moufarege A, Rodenberg C, Serum levels of androgens are
Cervantes C, Parra A (1983) Sleep Buch A, Purdie DW (2006) Efficacy higher in women with premenstrual
deprivation reduces circulating and safety of a testosterone patch irritability and dysphoria than in
androgens in healthy men. Arch for the treatment of hypoactive controls. Psychoneuroendocrinology
Androl 10:3337 sexual desire disorder in surgically 17:195204
menopausal women: a randomized, Farquhar CM, Sadler L, Harvey S,
Couzinet B, Meduri G, Lecce MG,
placebo-controlled trial. Menopause McDoudall J, Yazdi G, Meuli K
Young J, Brailly S, Loosfelt H,
13:387396 (2002) A prospective study of the
Milgrom E, Schaison G (2001) The
short-term outcomes of
postmenopausal ovary is not a Davison SL, Bell R, Donath S, Montalto
hysterectomy with and without
major androgen-producing gland. JG, Davis SR (2005) Androgen
oophorectomy. Aust N Z J Obstet
J Clin Endocr Metab 86:50605066 levels in adult females: changes with
Gynaecol 42:197204
Crilly RG, Marshall DH, Nordin BE age, menopause and oophorectomy.
(1979) The effect of age on plasma J Clin Endocr Metab 90:38473853 Fernandez-Guasti A, Kruijver FPM,
Fodor M, Swaab DF (2000) Sex
androstenedione concentration in Davison SL, Bell RJ, LaChina M,
differences in the distribution of
oophorectomised women. Clin Holden SL, Davis SR (2009) The
androgen receptors in the human
Endocrinol 10:199201 relationship between self-reported
hypothalamus. J Comp Neurol
sexual satisfaction and general
Dabbs JM Jr, Ruback RB, Frady Rl, 425:422435
Hopper CH, Sgoutas DS (1988) well-being in women. J Sex Med
Saliva testosterone and criminal 6:26902697 Finley SK, Kritzer MF (1999) Immuno-
reactivity for intracellular
violence among women. Pers Indiv Derogatis L, Rust J, Golombok S,
androgenreceptors in identified
Differ 9:269275 Bouchard C, Nachtigall L,
subpopulations of neurons,
Darney PD (1995) The androgenicity Rodenberg C, Kuznicki J,
astrocytes and oligodendrocytes in
of progestins. Am J Med 98 (Suppl McHorney CA (2004) Validation of
primate prefrontal cortex.
the profile of female sexual function
1A):104S110S J Neurobiol 40:446457
(PFSF) in surgically and naturally
Davidson JM, Carmago CA, Smith ER menopausal women. J Sex Marital Frenken J, Vennix P (1981) Sexuality
(1979) Effects of androgens on Ther 30:2536 Experience Scales Manual. Swets and
sexual behavior of hypogonadal Zeitlinger BV, Zeist, the
men. J Clin Endocr Metab Derogatis LR (1994) Symptom Netherlands
48:955958 Checklist-90-R: Administration,
Scoring, and Procedures Manual. Garratt A, Torgerson D, Wyness J, Hall
Davis AR, Castano PM (2004) Oral National Computer Systems Inc, M, Reid DM (1995) Measuring
contraceptives and libido in women. Minneapolis, MN sexual function in pre menopausal
Annu Rev Sex Res 15:297320 women. Br J Obstet Gynaecol
Derogatis LR (1997) The Derogatis 102:311316
Davis S, Papalia MA, Norman RJ, Interview for Sexual Functioning
ONeill S, Redelman M, Williamson (DISF/DISFSR): an introductory Gladue BA (1991) Aggressive
M, Stuckey BG, Wlodarczyk J, report. J Sex Mar Ther 231:291304 behavioral characteristics,
Gardner K, Humberstone A (2008) hormones, and sexual orientation in
Safety and efficacy of a testosterone Dow MGT, Gallagher J (1989) men and women. Aggress Behav
metered-dose transdermal spray for A controlled study of combined 17:313326
treating decreased sexual hormonal and psychological
treatment for sexual Goldstat R, Briganti E, Tran J, Wolfe R,
satisfaction in pre-menopausal Davis SR (2003) Transdermal
women: a randomized trial. Ann unresponsiveness in women. Brit
J Clin Psychol 28:201212 testosterone therapy improves well-
Intern Med 148:569577 being, mood and sexual function in

117
Chapter 5: The behavioral correlates of testosterone

pre-menopausal women. self-esteem. J Pers Soc Psychol gradually increasing doses of


Menopause 10:390398 60:895910. testosterone cypionate [published
Gooren LJG (1988) Hypogonadotropic Hedricks CA (1994) Female sexual correction appears in Drug Alcohol
hypogonadal men respond less well activity across the human menstrual Depend 1998; 50:255]. Drug Alcohol
to androgen substitution treatment cycle: a biopsychosocial approach. Depend 40:7379
than hypergonadotropic Annu Rev Sex Res 5:122172 Kraemer HC, Becker HB, Brodie HTH,
hypogonadal men. Arch Sex Behav Hines M (2004) Brain Gender. Oxford Doering CH, Moos RH, Hamburg
17:265270 University Press, Oxford DA (1976) Orgasmic frequency and
Gorski RA (2000) Sexual plasma testosterone levels in normal
Huesmann LR, Eron LD (1989) human males. Arch Sex Behav 5:125
differentiation of the nervous Individual differences and the trait
system. In: Kandel ER, Schwartz JH, of aggression. Europ J Personal Kwan M, Greenleaf WJ, Mann J, Crapo
Jesell TM (eds) Principles of Neural 3:95106 L, Davidson JM (1983) The nature
Science, 4th edn. McGraw-Hill, New of androgen action on male
York, pp 11311148 Hyde JS (1984) How large are gender
sexuality: a combined laboratory-
differences in aggression?
Graham CA (2010) The DSM self-report study on hypogonadal
A developmental meta-analysis.
diagnostic criteria for Female Sexual men. J Clin Endocr Metab
Develop Psychol 20:722736
Arousal Disorder. Arch Sex Behav 57:557562
39:240255 Janaud A, Rouffy J, Upmalis D, Dain
Laan E, Everaerd W (1995)
M-P (1992) A comparison of lipid
Graham CA, Bancroft J, Doll HA, Determinants of sexual arousal:
and androgen metabolism with
Greco T, Tanner A (2007) Does psychophysiological theory and
triphasic oral contraceptive
oral-contraceptive-induced data. Annu Rev Sex Res 6:3276
formations containing norgestimate
reduction in free testosterone or levonorgestrel. Acta Obstet Lange JD, Brown WA, Wincze JP,
adversely affect the sexuality Gynecol Scand Suppl 156:3338 Zwick W (1980) Serum testosterone
or mood of women? concentration and penile
Psychoneuroendocrinology Jung-Hoffman C, Kuhl H (1987)
Divergent effects of two low-dose tumescence changes in men. Horm
32:246255 Behav 14:267
oral contraceptives on sex
Gray A, Jackson DN, McKinlay JB hormone-binding globulin and free Leibenluft E, Schmidt PJ, Turner EH,
(1991) The relation between testosterone. Amer J Obs Gynecol Danaceau MA, Ashman SB, Wehr
dominance, anger, and hormones in 156:199203 TA, Rubinow DR (1997) Effects of
normally aging men: results from leuprolide-induced hypogonadism
Kelly DD (1991) Sexual differentiation
the Massachusetts male aging study. and testosterone replacement on
of the nervous system. In: Kandel
Psychosom Med 53:375385 sleep, melatonin and prolactin
ER, Schwartz JH, Jessell TM (eds)
Hamilton M (1960) A rating scale for Principles of Neural Science. secretion in men. J Clin Endocr
depression. J Neurol Neurosurg Appleton & Lange, Norwalk, pp Metab 82:32033207
Psychiatry 23:5662 959973 Liu PY, Yee B, Wishart SM, Jimenez M,
Hamilton M (1967) Development of a King SR (2008) Emerging roles Jung DG, Grunstein RR,
rating scale for primary depressive for neurosteroids in sexual Handelsman DJ (2003) The short-
illness. Br J Soc Clin Psychol behavior and function. Andrologia term effects of high-dose
6:278296 29:524533 testosterone on sleep, breathing, and
function in older men. J Clin Endocr
Harris JA (1999) Review and Komisaruk BR, Sansone G (2003) Metab 88:36053613
methodological considerations in Neural pathways mediating vaginal
research on testosterone and function: the vagus nerve and spinal Lloyd EA (2005) The Case of the Female
aggression. Aggress Viol Behav cord oxytocin. Scandinav J Psychol Orgasm: Bias in the Science of
4:273291 44:241250 Evolution. Harvard University
Press, Cambridge, MA
Harris JA, Rushton JP, Hampson E, Komisaruk BR, Frangos E, Whipple B
Jackson DN (1996) Salivary (2011) Hysterectomy improves Luboshitzky R, Aviv A, Hefetz A, Herer
testosterone and self-report sexual response? Addressing a P, Shen-Orr Z, Lavie L, Lavie P
aggressive and prosocial personality crucial omission in the literature. (2002) Decreased pituitary-gonadal
characteristics in men and women. J Minim Invas Gyn 18:288295 secretion in men with obstructive
Aggress Behav 22:313331 Kouri EM, Lukas SE, Pope HG Jr, Oliva
sleep apnea. J Clin Endocr Metab
87:33943398
Heatherton TF, Polivy J (1991) PS (1995) Increased aggressive
Development and validation responding in male volunteers Luisi M, Franchi F (1980) Double-blind
of a scale for measuring state following the administration of group comparative study of

118
Chapter 5: The behavioral correlates of testosterone

testosterone undecanoate and erections: an overview. J Sex Med for Boys. Report No. 6. University of
mesterolone in hypogonadal 2:771784 Bergen, Bergen, Norway
male patients. J Endocrinol Invest Mushayandebvu T, Castracane VD, Olweus D, Mattsson A, Schalling D,
3:305308 Gimpel T, Adel T, Santoro N (1996) Low H (1980) Testosterone,
Manetti GJ, Honig SC (2010) Update Evidence for diminished mid-cycle aggression, physical, and
on male hormonal contraception: is ovarian androgen production in personality dimensions in normal
the vasectomy in jeopardy? Internat older reproductive aged women. adolescent males. Psychosom Med
J Impot Res 22:159170 Fert Steril 65:721723 42:253269
Mathews A, Whitehead A, Kellett J Myers LS, Dixen J, Morrissette D, Olweus D, Mattsson A, Schalling D,
(1983) Psychological and hormonal Carmichael M, Davidson JM (1990) Low H (1988) Circulating
factors in the treatment of female Effects of estrogen, androgen, and testosterone levels and
sexual dysfunction. Psychol Med progestin on sexual aggression in adolescent males: a
13:8392 psychophysiology and behavior in causal analysis. Psychosom Med
postmenopausal women. J Clin 42:253269
McNair DM, Lorr M, Droppleman LF
Endocr Metab 70:11241131 Orentreich N, Brind JL, Rizer RL,
(1992) EdITS Manual for the Profile
of Mood States. Educational and Nathorst-Bs J, von Schoultz B, Vogelman JH (1984) Age changes
Industrial Testing Service, San Carlstrm K (1993) Elective and sex differences in serum
Diego ovarian removal and estrogen dehydroepiandrosterone sulfate
replacement therapy effects on concentrations throughout
Meyer-Bahlburg HFL, Baker SW, sexual life, psychological adulthood. J Clin Endocr Metab
Dolezal C, Carlson AD, Obeid JSD, wellbeing and androgen status. 59:551555
New MI (2003) Long-term outcome J Psychosom Obstet Gynaecol
in congenital adrenal hyperplasia: Overall JE, Gorham DR (1962) The
14:283293 Brief Psychiatric Rating Scale.
gender and sexuality.
Endocrinologist 13:227232 National Institute for Health and Psychol Res 10:799812
Clinical Excellence (NICE) (2005) Panjari M, Davis SR (2010) DHEA
Michael RP, Rees HD, Bonsall RW Long-Acting Reversible
(1989) Sites in the male primate for postmenopausal women: a
Contraception: The Effective and review of the evidence. Maturitas
brain at which testosterone acts as Appropriate Use of Long-Acting
an androgen. Brain Res 502:1120 66:172179
Reversible Contraception. RCOG
Miller KK, Sesmilo G, Schiller A, Press, London Parent AS, Teilmann G, Juul A,
Schoenfeld D, Burton S, Klibanski A Skakkebaek NE, Toppari J,
Neave N, Wolfson S (2003) Bourguignon J-P (2003) The timing
(2001) Androgen deficiency in Testosterone, territoriality, and the
women with hypopituitarism. J Clin of normal puberty and the age limits
home advantage. Physiol Behav of sexual precocity: variations
Endocr Metab 86:561567 78:269275 around the world, secular trends,
Miller KK, Biller BM, Beauregard C, OCarroll RE, Shapiro C, Bancroft J and changes after migration. Endocr
Lipman JG, Jones J, Schoenfeld D, (1985) Androgens, behaviour and Rev 24:668693
Sherman JC, Swearingen B, Loeffler nocturnal erections in hypogonadal Park K, Kang HK, Seo JJ, Kim HJ, Ryu
J, Klibanski A (2006) Effects of men: the effect of varying the SB, Jeong GW (2001) Blood-
testosterone replacement in replacement dose. Clin Endocrinol oxygenation level-dependent
androgen-deficient women with 23:527538 functional magnetic resonance
hypopituitarism: a randomized,
OConnor DB, Archer J, Wu FCW imaging for evaluating cerebral
double-blind, placebo-controlled
(2001) Measuring aggression: self- regions of female sexual arousal
study. J Clin Endocr Metab
reports, partner reports and response. Urology 57:11891194
91:16831690
responses to provoking scenarios. Parmeggiana PL, Morrison AR (1990)
Minto CL, Liao KL-M, Conway GS, Aggress Behav 27:79101 Alterations in autonomic functions
Creighton SM (2003) Sexual
OConnor DB, Archer J, Wu FCW during sleep. In: Loewy AD, Spyer
function in women with complete
(2004) Effects of testosterone on KM (eds) Central Regulation of
androgen insensitivity syndrome.
mood, aggression, and sexual Autonomic Functions. Oxford
Fert Steril 80:157164
behavior in young men: a double- University Press, New York,
Money J, Ehrhardt A (1972) Man and blind, placebo-controlled, cross- pp 367386
Woman: Boy and Girl. Johns over study. J Clin Endocri Metab Perrin JS, Herv PY, Leonard G, Perron
Hopkins Press, Baltimore 89:28372845 M, Pike GB, Pittiot A, Richer L,
Montorsi F, Oettel M (2005) Olweus D (1975) Development of a Veillette S, Pausova Z, Paus T
Testosterone and sleep-related Multi-Faceted Aggression Inventory (2008) Growth of white matter in

119
Chapter 5: The behavioral correlates of testosterone

the adolescent brain: role of Roselli CE, Klosterman S, Resko JA major and minor depression. Arch
testosterone and androgen receptor. (2001) Anatomic relationships Gen Psychiat 62:154162
J Neurosci 28:95199524 between aromatase and Schreiner-Engel P, Schiavi RC, White
Pope HG Jr, Kouri EM, Hudson JI androgen receptor mRNA D, Ghizzani A (1995) Low sexual
(2000) Effects of supraphysiological expression in the hypothalamus desire in women: the role of
doses of testosterone on mood and and amygdala of adult male reproductive hormones. Horm
aggression in normal men. Arch cynomolgus monkeys. J Comp Behav 23:221234
Gen Psychiatry 57:133140 Neurol 439:208223
Seigel JM (1986) The multi-
Puy L, MacLusky NJ, Becker L, Karsan Rubin H, Henson D, Falvo R, High R dimensional anger inventory.
N, Trachtenberg J, Brown TJ (1995) (1979) The relationship between J Personality Soc Psych 51:191200
Immuno-cytochemical detection of mens endogenous levels of
testosterone and their penile Sherwin BB (1988) A comparative
androgen receptor in human
responses to erotic stimuli. Behav analysis of the role of androgen in
temporal cortex: characterization
Res Ther 17:305 human male and female sexual
and application of polyclonal
behavior: behavioral specificity,
androgen receptor antibodies in Salmimies P, Kockott G, Pirke KM, critical thresholds, and sensitivity.
frozen and paraffin-embedded Vogt HJ, Schill WB (1982) Effects of Psychobiology 16:416425
tissues. J Steroid Biochem Mol Biol testosterone replacement on sexual
55:197209 behavior in hypogonadal men. Arch Sherwin BB, Gelfand MM (1985a) Sex
Sex Behav 11:345353 steroids and affect in the surgical
Raboch J, Starka L (1973) Reported
menopause: a double-blind, cross-
coital activity of men and levels of Sandblom RE, Matsumoto AM, Scoene over study. Psychoneuroendocrinol
plasma testosterone. Arch Sex Behav RB, Lee KA, Giblin EC, Bremner 10:325335
2:309 WJ, Pierson DJ (1983) Obstructive
sleep apnoea induced by Sherwin BB, Gelfand MM (1985b)
Radloff LS (1977) The CES-D Scale.
testosterone administration. N Engl Differential symptom response to
A self-report depression scale for
J Med 308:508510 parenteral estrogen and/or
research in the general population.
androgen administration in the
App Psych Meas 1:385401 Sanders SA, Graham CA, Bass J, surgical menopause. Am J Obstet
Rathus SA (1973) A 30-item schedule Bancroft J (2001) A prospective Gynecol 151:153160
for assessing assertive behavior. study of the effects of oral
Behav Ther 4:398406 contraceptives on sexuality and Sherwin BB, Gelfand MM, Brender W
well-being and their relationship to (1985) Androgen enhances sexual
Redout J, Stolru S, Pugeat M, Costes motivation in females: a
discontinuation. Contraception
N, Lavenne F, Le Bars D, Dechaud prospective, crossover study of sex
64:5158
H, Cinotti L, Pujol JF (2005) Brain steroid administration in the
processing of visual sexual stimuli Sarkey S. Azcoita I, Garcia-Segura LM, surgical menopause. Psychosom
in treated and untreated Garcia-Ovejero D, DonCarlos LL Med 47:339351
hypogonadal patients. (2008) Classical androgen receptors
Psychoneuroendocrinology, in non-classical sites in the brain. Shifren JL, Braunstein GD, Simon JA,
30:461482 Horm Behav 53:753764 Casson PR, Buster JE, Redmond
GP, Burki RE, Ginsberg ES, Rosen
Riley A, Riley E (2000) Controlled Sarrieau A, Mitchell JB, Lal S, Olivier RC, Leiblum SR, Caramelli KE,
studies on women presenting with A, Quirion E, Meaney MJ (1990) Mazer NA (2000) Transdermal
sexual drive disorder: I. Endocrine Androgen binding sites in human testosterone treatment in women
status. J Sex Marital Ther temporal cortex. with impaired sexual function after
26:269283 Neuroendocrinology 51:713716 oophorectomy. N Engl J Med
Roger M, Nahoul K, Scholler R, Bagrel Schmidt PJ, Berlin KL, Danaceau MA, 343:682688
D (1980) Evolution with ageing of Neeren A, Haq MA, Roca CA, Shifren JL, Davis SR, Moreau M,
four plasma androgens in Rubinow DR (2004) The effects of Waldbaum A, Bouchard C,
postmenopausal women. Maturitas pharmacologically induced Derogatis L, Derzco C, Bearnson P,
2:171177 hypogonadism on mood in healthy Kakos N, ONeill S, Levine S,
Rommerts FFG (1990) Testosterone: men. Arch Gen Psychiat Wechselman K, Buch A, Rodenberg
an overview of biosynthesis, 61:9971004 C, Kroll R (2006) Testosterone
transport, metabolism and action. Schmidt PJ, Daly RC, Bloch M, Smith patch for the treatment of
In: Nieschlag E, Behre HM (eds) MJ, Danaceau MA, Simpson St.CL, hypoactive sexual desire disorder in
Testosterone: Action, Deficiency, Murphy JH, Haq N, Rubinow DR naturally menopausal women:
Substitution. Springer-Verlag, (2005) Dehydroepiandrosterone results from the INTIMATE NM1
Berlin, pp 122 monotherapy in midlife-onset Study. Menopause 13:770779

120
Chapter 5: The behavioral correlates of testosterone

Shores MM, Moceri VM, Sloan KL, Taylor JF, Rosen RC, Leiblum SR impaired sperm production, and
Alvin M, Matsumoto MD, Kivlahan (1994) Self-report assessment of male infertility. J Clin Endocr Metab
DR (2005) Low testosterone levels female sexual function: 82:37773782
predict incident depressive illness in psychometric evaluation of the Vague J (1983) Testicular feminization
older men: effects of age and Brief Index of Sexual Functioning syndrome: an experimental model
medical morbidity. J Clin Psychiat for Women. Arch Sex Behav for the study of hormone action on
66:714 23:627643 sexual behavior. Horm Res 18:6268
Simmons ZL, Roney JR (2011) Teplin V, Vittinghoff E, Lin F, Van der Pahlen B, Lindman R, Sarkola
Variation in CAG repeat Learman LA, Richter HE, T, Mkisalo H, Eriksson CJP (2002)
length of the androgen receptor Kuppermann M (2007) An exploratory study on self-
gene predicts variables associated Oophorectomy in premenopausal evaluated aggression and
with intrasexual competitiveness women. Health-related quality of androgens in women. Aggress Behav
in human males. Horm Behav life and sexual functioning. Obstet 28:273280
60:306312 Gynecol 109:347354
Van der Vange N, Blankenstein MA,
Simon J, Braunstein G, Nachtigall L, Thorneycroft IH, Stanczyk FZ, Kloosterboer HJ, Haspels AA,
Utian W, Katz M, Miller S, Bradshaw KD, Ballagh SA, Nichols Thijssen JH (1990) Effects of seven
Waldbaum A, Bouchard C, Derzko M, Weber ME (1999) Effect of low- low-dose combined oral
C, Buch A, Rodenberg C, Lucas J, dose oral contraceptives on contraceptives on sex hormone
Davis S (2005) Testosterone patch androgenic markers and acne. binding globulin, corticosteroid
increases sexual activity and desire Contraception 60:255262 binding globulin, total and free
in surgically menopausal women testosterone. Contraception
Thornton JW (2001) Evolution of
with hypoactive sexual desire 41:345352
vertebrate steroid receptors from an
disorder. J Clin Endocrinol Metab
ancestral estrogen receptor by Van Goozen SHM, Cohen-Kettenis PT,
90:52265233
ligand exploitation and serial Gooren LJG, Frijda NH, Van de Poll
Simpson JA, Gangestad SW (1991) genome expansions. PNAS NE (1995) Gender differences in
Individual differences in 98:56715676 behaviour: activating effects of
sociosexuality evidence for cross-sex hormones.
convergent and discriminant Tricker R, Casaburi R, Storer TW,
Clevenger B, Berman N, Shirazi A, Psychoneuroendocrinology
validity. J Pers Soc Psychol 20:343363
60:870883 Bhasin S (1996) The effect of
supraphysiological doses of Vermeersch H, TSjoen G, Kaufman
Skakkebaek NE, Bancroft J, Davidson testosterone on angry behavior in JM, Vincke J, Van Houtte M (2010)
DW, Warner P (1981) Androgen healthy eugonadal men a clinical Testosterone, androgen receptor
replacement with oral testosterone research center study. J Clin Endocr gene CAG repeat length, mood and
undecanoate in hypogonadal men: a Metab 81:37543758 behaviour in adolescent males.
double blind controlled study. Clin Europ J Endocrinol 163:319328
Endocrinol 14:4961 Tuiten A, Laan E, Panhuysen G,
Everaerd W, de Haan E, Walker PA (1978) The role of
Stuart FM, Hammond DC, Pett MA Koppeschaar H, Vroon P (1996) antiandrogens in the treatment of
(1987) Inhibited sexual desire in Discrepancies between genital sex offenders. In: Qualls CB, Wincze
women. Arch Sex Behav 16:91106 responses and subjective sexual JP, Barlow DH (eds) The Prevention
Su TP, Pagliaro M, Schmidt PJ, Pickar function during testosterone of Sexual Disorders. Plenum, New
D, Wolkowitz O, Rubinow DR substitution in women with York, pp 117136
(1993) Neuropsychiatric effects of hypothalamic amenorrhea.
Weber B, Lewicka S, Deuschle M, Colla
anabolic steroids in male normal Psychosom Med 58:234241
M, Heuser I (2000) Testosterone,
volunteers. JAMA 269:27602764 Tuiten A, Van Honk J, Koppeschaar H, androstenedione and
Sulcov J, Hill M, Hampl R, Strka L Bernaards C, Thijssen J, Verbaten R dihydrotestosterone concentrations
(1997) Age and sex related (2000) Time course of effects of are elevated in female patients with
differences in serum levels of testosterone administration on major depression.
unconjugated sexual arousal in women. Arch Gen Psychoneuroendocrinology
dehydroepiandrosterone and its Psychiat 57:149153 25:765771
sulphate in normal subjects. Tut TG, Ghadessy FJ, Trifiro MA, Weinberger DR, Elvevag B, Giedd JN
J Endocrinol 154:5762 Pinsky I, Yong EL (1997) Long (2005) The Adolescent Brain:
Symons D (1979) The Evolution of polyglutamine tracts in the A Work in Progress. The National
Human Sexuality. Oxford androgen receptor are associated Campaign to Prevent Teen
University Press, New York with reduced transactivation, Pregnancy, Washington, DC.

121
Chapter 5: The behavioral correlates of testosterone

Available at: www.teenpregnancy. Yates WR, Perry PJ, MacIndoe J, constructs. Europ J Endocrinol
org/resources/reading/pdf/BRAIN. Holman T, Ellingrod V (1999) 144:183197
pdf (accessed Jan 16, 2012) Psychosexual effects of three
Zitzmann M, Nieschlag E (2003) The
Westberg L, Baghaei F, Rosmond R, doses of testosterone cycling in
CAG repeat polymorphism within
Hellstrand M, Landn M, Jansson normal men. Biol Psychiat
the androgen receptor gene and
45:254260
M, Holm G, Bjrntorp P, Eriksson E maleness. Int J Androl 26:7683
(2001) Polymorphisms of the Young RC, Biggs JT, Ziegler VE, Meyer
androgen receptor gene and the Zitzmann M, Faber S, Nieschlag E
DA (1978) A rating scale for mania:
estrogen receptor b gene are (2006) Association of specific
reliability, validity, and sensitivity.
symptoms and metabolic risks
associated with androgen levels in Br J Psychiat 133:429435
women. J Clin Endocr Metab with serum testosterone in older
86:25622568 Zhou J, Hofman MA, Gooren LJG, men. J Clin Endocr Metab
Swaab DF (1995) A sex difference in 91:43354343
Wisniewski AB, Migeon CJ, Meyer- the human brain and its relation to
Bahlburg HFL, Gearhart JP, Zumoff B, Strain GW, Miller LK,
transexuality. Nature 378:6870
Berkowitz GD, Brown TR (2000) Rosner W (1995) Twenty-four-hour
Complete androgen insensitivity Zitzmann M, Nieschlag E (2001) mean plasma testosterone
syndrome: long-term medical, Testosterone levels in healthy men concentration declines with age in
surgical and psychosexual outcome. and the relation to behavioural and normal premenopausal women.
J Clin Endocr Metab 85:26642669 physical characteristics: facts and J Clin Endocr Metab 80:14291430

122
Chapter
The role of testosterone in spermatogenesis

6 Liza ODonnell and Robert I. McLachlan

6.1 Introduction 123 6.5.3 Mechanisms of androgen action in the


6.2 Organization and kinetics of testis 135
spermatogenesis 124 6.5.4 Relationship between testicular androgen
6.2.1 Basic and common features 124 concentrations and spermatogenic
6.2.2 Species-specific features 126 progression 137
6.3 The hypothalamo-hypophyseal-testicular 6.6 Other endocrine factors in the control of
axis 126 spermatogenesis 138
6.4 Testosterone and spermatogenesis 127 6.6.1 Follicle-stimulating hormone and
6.4.1 Neonatal androgen secretion 127 spermatogenesis 138
6.4.2 Role of androgens in the initiation and 6.6.2 Androgen and follicle-stimulating
maintenance of hormone; cooperative effects on
spermatogenesis 128 spermatogenesis 140
6.4.3 Adult spermatogenesis: maintenance 6.6.3 Aromatization of testosterone to estradiol; role
and reinitiation 129 in spermatogenesis 143
6.5 Androgen action on spermatogenesis 130 6.7 Implications for treatment of secondary
6.5.1 Testicular androgen production and hypogonadism and for male hormonal
metabolism 130 contraception 144
6.5.2 Testicular androgen receptor and sites 6.8 Key messages 144
of androgen action 132 6.9 References 145

6.1 Introduction transgenic models have also been utilized extensively


Testosterone production and action is critical for and provide important information on the sites and
male fertility. An absence of androgen signaling mechanisms of androgen action in spermatogenesis.
during fetal life results in failure of the urogenital Luteinizing hormone from the pituitary stimulates the
tract to virilize, inhibition of testicular descent, abnor- Leydig cells of the testis to produce testosterone, from
malities in accessory organs, such as the epididymis, fetal life through to adulthood. Testosterone is essential
prostate and seminal vesicles, and an inability to for many aspects of spermatogenesis, including meiosis
produce sperm. This chapter focuses on the role of and differentiation of haploid germ cells (a process
testosterone in the initiation and maintenance of known as spermiogenesis). Androgen action is primar-
germ cell development, known as spermatogenesis. ily mediated by ARs within Sertoli cells; however, AR-
Much of what we know about testosterone and mediated action on Leydig cells and peritubular myoid
spermatogenesis is based on studies in which testoster- cells is also important. The specific molecular pathways
one levels are experimentally manipulated in rodents, of androgen action are not yet well characterized, with
non-human primates and men. In recent times, both genomic and non-genomic pathways thought

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

123
Chapter 6: The role of testosterone in spermatogenesis

to be involved. While androgen-dependent genes recombination occurs to ensure the genetic diversity
have been discovered, there are surprisingly few. of the gametes. Cells then undergo two meiotic div-
Evidence is emerging that androgens may modulate isions in rapid succession to produce haploid round
spermatogenesis by the coordination of transcriptional spermatids. These spermatids undergo a major and
and translational events, including the regulation of complex morphological, structural and functional
small RNA species that modulate protein expression. maturation and development process (spermiogen-
In this chapter, we review the current information esis), resulting in the production of mature elongated
on the roles of testosterone in the initiation and main- spermatids that are released into the seminiferous
tenance of spermatogenesis in rodents, monkeys and epithelium (spermiation), yet these cells do not
men. While testosterone has independent effects on exhibit progressive motility but are capable of fertil-
spermatogenesis, it also cooperates with FSH to facili- ization following injection into oocytes in vitro.
tate quantitatively normal sperm production, and thus Germ cell development occurs in close contact with
we will briefly cover the role of FSH and its synergy the somatic cells of the epithelium, the Sertoli cells (Fig.
with testosterone. Because testosterone can be metab- 6.1). These cells possess highly specialized cytological
olized to estradiol in the testis, and estrogenic actions and structural features enabling them to functionally
can influence male fertility, we will briefly review the and physically support the development of germ cells as
role of estrogen in spermatogenesis. Our primary they move within the epithelium from the basement
focus will be on the sites and mechanisms of androgen membrane (spermatogonia) to the luminal edge prior
action that are essential for germ cell development. to the release of sperm. Sertoli cells divide during the
This information will be explored in terms of the fetal and early pubertal period to produce a fixed popu-
clinical importance of androgens in normal and dis- lation of cells that determines the size and sperm output
ordered gonadal development, how deficiency states of the adult testis (De Franca et al. 1995). At the end of
are best managed to restore fertility, and what strat- their proliferative period, Sertoli cells undergo a ter-
egies can be used for the withdrawal of testicular minal differentiation and form a series of tight and
androgenic support for the purposes of contraception. occluding junctions that provide the basis of the
bloodtestis barrier, an exclusion barrier that allows
the Sertoli cell to determine the microenvironment of
6.2 Organization and kinetics the meiotic and post-meiotic germ cells.
of spermatogenesis Among mammals, germ cell development is not
randomly distributed within the seminiferous epithe-
6.2.1 Basic and common features lium, but is arranged in strictly defined cellular asso-
The testis contains the seminiferous tubules and ciations (Fig. 6.1; reviewed in de Kretser and Kerr
interstitial tissue, enclosed by a capsule called the 1988; Russell et al. 1990). A particular association of
tunica. Male gamete development, or spermatogen- germ cells is referred to as a stage, and the number of
esis, occurs within the seminiferous tubules (Fig. 6.1). stages of spermatogenesis in a particular species is
The interstitial tissue contains the steroidogenic Ley- defined by the number of morphologically recogniz-
dig cells which produce testosterone, as well as the able germ cell associations. Every stage is thought to
blood and lymphatic vessels that are essential for the derive from one stem cell and hence represents a cell
movement of hormones and nutrients into, and out of, clone, with intercellular bridges remaining that allow
the testis, and a population of resident macrophages. continued cellular communication (Ren and Russell
The seminiferous tubules are surrounded by peritubu- 1991). The seminiferous epithelium is staged based
lar myoid cells and layers of basement membrane. upon the morphology of the developing acrosome in
Spermatogenesis comprises the development of spermatids (Clermont and Leblond 1955), and the
sperm from spermatogonia. Spermatogonial stem number of spermatogenic stages varies between
cells divide and self-renew before becoming commit- species. For example, there are 12 stages in mice, 14
ted to differentiation. Differentiating spermatogonia in rats and 6 in humans (Russell et al. 1990). Dividing
undergo a series of mitoses to amplify the pool of cells the spermatogenic process into stages is critical
that enter meiosis. Once primary spermatocytes enter because many processes and actions occur physiolo-
meiosis, they undergo a long (several weeks) prophase gically in a stage-specific manner. Conversely, dis-
where chromosomes are replicated and homologous turbances of spermatogenesis imposed by endocrine

124
Chapter 6: The role of testosterone in spermatogenesis

Epididymis Fig. 6.1 The organization of the


testis and seminiferous epithelium.
Diagram of the testis and epididymis is
Efferent shown, along with a cross-section of rat
ductules testis (bar = 50 mm). The seminiferous
tubules have a well-defined lumen, into
Lumen Leydig cells which the mature sperm are released
Vas
deferens Rete prior to their passage to the epididymis
via the rete testis and then efferent
Interstitium ductules. The interstitium contains the
steroidogenic Leydig cells that produce
testosterone, as well as the blood and
lymphatic tissues which allow for the
exchange of hormones and nutrients
Testis between the testis and the circulation.
A cross-section through adult rat
seminiferous epithelium shows a Sertoli
cell (with prominent nucleolus) with its
Seminiferous cytoplasmic extensions outlined as they
tubule encompass germ cells at various stages of
development. This particular cross-section
shows the typical cellular associations
seen at stage VII of the rat spermatogenic
eST cycle. Basement membrane and
peritubular myoid cells encircle the
outside of the tubule. Abbreviations:
Sg, spermatogonia; Pl, preleptotene
spermatocyte; PS, pachytene
rST spermatocyte; rST, round spermatid;
eST, elongated spermatid.

PS

Pl
Sg
Basement
membrane

Sertoli cell
Peritubular
myoid cell

Seminiferous
epithelium

deficiencies or exposure to toxicants at least initially Whereas it was previously believed that the Sertoli
frequently manifest in a stage-specific pattern. Each cells govern the kinetics of the spermatogenic process,
stage of spermatogenesis exhibits species-specific it is now thought that this ability resides within the
timing, and the length of time for a complete cycle germ cell itself. Germ cell transplantation from rat to
(i.e. all stages in succession) also differs; for example, mouse testes revealed that rat germ cell development
the duration of one cycle is 79 days for the mouse, could be supported by mouse Sertoli cells and that the
1214 days for the rat, and 16 days for man (Russell transplanted rat germ cells developed according to the
et al. 1990). The different kinetics of spermatogenesis kinetics of rat spermatogenesis (Franca et al. 1998).
result in the time taken for spermatogonia to develop This suggests that germ cells develop according to
into sperm differing; e.g. 35 days in mice and 64 days their own predetermined timing, which will then
in men. The kinetics of spermatogenesis are unlikely to influence stage-related morphological and functional
be affected by gonadotropin manipulation (reviewed in changes within the Sertoli cells. However, stage-
ODonnell et al. 2006). related changes occurred in embryonic Sertoli cells

125
Chapter 6: The role of testosterone in spermatogenesis

in the absence of mature germ cells (Timmons et al. Type Ap spermatogonia undergo mitosis and provide
2002), indicating that these cells possess an ability to daughter cells to enter the spermatogenic process and
undergo time-dependent functional changes well to renew the spermatogonial stem cell population.
before spermatogenesis is established. Type Ad spermatogonia rarely divide in the intact
Evaluation of the spermatogenic process can be testis and are considered to replenish the Ap sperma-
qualitative or quantitative. Qualitatively normal togonial population in the case of severe spermatogo-
spermatogenesis refers to the presence of all germ cell nial depletion; e.g. following testicular irradiation
types and spermatogenic stages; whereas quantita- (van Alphen et al. 1989). Conversely, there may be a
tively normal spermatogenesis refers to the produc- transition of Ap into Ad spermatogonia as a protective
tion of all germ cell types in normal numbers. This mechanism during various onslaughts, such as gona-
distinction is very important for the discussion of the dotropin suppression or irradiation (van Alphen et al.
relative role of testosterone and FSH in spermatogenesis 1988; ODonnell et al. 2001a).
and for the assessment of toxic actions on spermatogen- Among rodents, a tubule cross-section is occupied
esis. Elongated spermatids possess tightly compacted by a single spermatogenic stage (single-stage arrange-
heads which, unlike all other testicular cells, are ment); however, in primates more complex, multi-
resistant to homogenization in detergent-based stage arrangements are seen (Wistuba et al. 2003).
buffers. Accordingly the number of homogeniza- In New World monkeys, hominoids and humans,
tion-resistant spermatids per testis has long been tubules are predominantly multi-stage but are single
used to quantitatively assess spermatogenesis. The stage in macaques and intermediate in baboon. The
use of stereology to accurately enumerate specific human multi-stage arrangement has been suggested
germ cell populations in thick (25 mm) sections of to derive from a helical arrangement of spermato-
testis along with Sertoli cells (Wreford 1995) allows genic stages (Schulze and Rehder 1984), but this
an assessment of the kinetics of spermatogenic view has also been challenged (Johnson et al. 1996).
response to various experimental settings, and facili- Alternatively, it might be the size of each clone (see
tates a better understanding of how specific germ Section 6.2.1) that determines whether or not a par-
cells and cellular processes respond to endocrine ticular spermatogenic stage entirely occupies a tubule
manipulation. cross-section (Wistuba et al. 2003).

6.2.2 Species-specific features 6.3 The hypothalamo-hypophyseal-


Although the spermatogenic process has many testicular axis
common features, substantial differences must also The hypothalamic-pituitary-testicular (HPT) axis
be kept in mind when considering its hormonal regu- ensures the establishment and maintenance of testicular
lation. For the purpose of this chapter, the discussion function, specifically androgen secretion (for virilization)
of species-specific aspects is largely confined to a and spermatogenesis (for fertility). Gonadotropin-
comparison between rodents (mouse, rat, hamster) releasing hormone is secreted in a pulsatile fashion
and primates (non-human primates and man). from hypothalamic neurons into the hypophyseal
The system of spermatogonial renewal is quite portal circulation to stimulate the synthesis and
different between rodents and primates. Rodent stem release of the gonadotropin hormones, LH and FSH
spermatogonial development is well described, and from the pituitary (Fig. 6.2). Luteinizing hormone
several generations of differentiating and dividing stimulates testicular Leydig cells to synthesize and
type A spermatogonia exist prior to formation of type secrete testosterone. Testosterone levels within the
B spermatogonia (de Rooij and Grootegoed 1998). testis are exceedingly high (50100-fold serum levels)
In the primate, Ad(dark) spermatogonia are considered and act on the peritubular cells that surround the
to be the resting or reserve stem cell population, seminiferous tubules and, most importantly, on the
Ap(pale) spermatogonia are considered to be the pro- somatic Sertoli cells within their walls, thereby provid-
liferative spermatogonia, and B spermatogonia are ing critical support for spermatogenesis. It must be
considered to be committed to differentiation. The recognized that testosterone exerts pleiotropic effects
precise relationship between Ad and Ap spermato- on reproductive and non-reproductive tissues; ARs
gonia and their kinetics are still under investigation. are widely distributed, and androgens play major

126
Chapter 6: The role of testosterone in spermatogenesis

Hypothalamus cells via G-protein-mediated surface receptors to sup-


port spermatogenesis.
GnRH The secretion of GnRH and gonadotropin hor-
Pituitary mones is controlled by testicular steroid and protein
factors. Testosterone is the major steroid eliciting a
FSH LH negative feedback effect on LH at the pituitary level,
+ + but it predominantly acts at the hypothalamic level
Inhibin B T (Veldhuis et al. 1997; Fingscheidt et al. 1998). The
E regulation of FSH is more complex: in terms of
steroid feedback, estradiol appears to be the main
regulator in the adult male (Pitteloud et al. 2008).
However, the primary endocrine regulator of serum
Periphery FSH is the Sertoli cell product, inhibin B, which
Sertoli
+T T exerts a negative feedback signal at the gonadotrope
cell
Leydig level and reflects the adequacy of spermatogenesis
cell (Fingscheidt et al. 1998; Ramaswamy et al. 1998;
[iTT] = 50100-fold serum Pitteloud et al. 2008). Activin and follistatin are also
involved in FSH feedback regulation, but act more
as local regulators within the pituitary cell rather
Testis
than as endocrine factors (de Kretser et al. 2004;
Fig. 6.2 The hypothalamic-pituitary-testicular (HPT) axis in Bernard et al. 2010). Activins selectively stimulate
primates. Gonadotropin-releasing hormone (GnRH) is secreted in a
pulsatile fashion from hypothalamic neurons into the hypophyseal
FSH secretion, and follistatin binds to activin and
portal circulation to stimulate the synthesis and release of the presumably determines and regulates activin-associ-
gonadotropic hormones, luteinizing hormone (LH) and follicle- ated effects through this mechanism (McConnell
stimulating hormone (FSH) from the pituitary which have positive
effects (solid arrows) on testicular functions. Luteinizing hormone
et al. 1998). The physiological relevance of activin
acts on receptors within the Leydig cell plasma membrane to for spermatogenesis is strongly indicated by the
stimulate the synthesis and release of testosterone (T), resulting in an observation that over-expression of follistatin is
intratesticular testosterone concentration ([iTT]) approximately
50100-fold that of serum testosterone concentrations. Testosterone
associated with spermatogenic defects, reduced
acts on testicular somatic cells, including the Sertoli cells, to regulate testis size and reduced fertility in mice (Guo et al.
spermatogenesis, and is also secreted into the peripheral circulation 1998; de Kretser et al. 2001).
to maintain androgen-dependent functions. Testosterone exerts a
negative feedback effect (dashed arrows) on the pituitary release
of LH, but it predominantly acts via negative regulation of
hypothalamic GnRH secretion. The inhibitory effects of testosterone 6.4 Testosterone and
in the brain are mediated in part via its aromatization to estradiol (E).
Follicle-stimulating hormone acts directly on Sertoli cells of the testis spermatogenesis
to mediate spermatogenesis. Feedback regulation of FSH is
effected by the Sertoli cell product, inhibin B and by sex steroid, 6.4.1 Neonatal androgen secretion
predominately estradiol at the pituitary level.
Activation of the HPT axis occurs at three times
during human development: transiently during fetal
life, in the first three to six months of postnatal life
roles in metabolism, adipose tissue and hematopoietic, (Gendrel et al. 1980; Grumbach 2005) and then
muscle and brain function (Dankbar et al. 1995). continually from puberty. A distinct peak of testos-
Interference with these testosterone actions (or those terone synthesis and secretion occurs for variable
of its derivative steroid hormones) must be con- durations in the perinatal phase in other species,
sidered with strategies that manipulate testicular but the physiological significance of the accom-
androgen secretion or action. Testosterone is both a panying rise in testosterone production is not
hormone acting directly on androgen receptors but entirely clear.
also serves as a prohormone via its conversion to Inhibition of gonadotropin release by GnRH
other steroids; specifically 5a-reduction to DHT or antagonist treatment delayed puberty and testicular
aromatization to estradiol (Handelsman 2008). Fol- development in rats, but eventually fertility reverted
licle-stimulating hormone acts directly on Sertoli to normal (Kolho and Huhtaniemi 1989a; 1989b);

127
Chapter 6: The role of testosterone in spermatogenesis

while in primates no untoward effects on adult tes- with reduced/absent local iTT production (Kremer
ticular function and fertility were evident (Lunn et al. et al. 1995; Gromoll et al. 2000).
1997). These data cannot be taken to mean that the These data clearly demonstrate the ability of testos-
perinatal gonadotropin androgen surge has no role in terone to initiate the spermatogenic process; yet these
all non-human primates or humans. A deficiency in data cannot prove that testosterone is indispensable. For
the fetal and perinatal gonadotropin surge may have example, fertile eunuchs have atrophied Leydig cells
profound consequence in adulthood; for example in and complete spermatogenesis (Behre et al. 2010), yet
congenital GnRH deficiency, permanent effects on one cannot prove iTT levels are zero or that very low
penile size (micropenis) and a reduced Sertoli cell (unmeasurable) iTT levels are not providing support
proliferative phase that may limit adult testis size for limited spermatogenesis. This consideration may
and spermatogenic potential (Sharpe et al. 2003; also account for the observation that spermatogenesis
Grumbach 2005; Sykiotis et al. 2010). becomes established later in the life of the LH recep-
tor knock-out mouse (Zhang et al. 2003). One can
6.4.2 Role of androgens in the initiation conclude that spermatogenesis can be induced by
testosterone levels that are substantially lower than
and maintenance of spermatogenesis in normal puberty.
The LH-induced rise in intratesticular testosterone During pubertal initiation, testosterone also
(iTT) at puberty is the primary stimulus for the initi- stimulates GH secretion and GH-dependent growth
ation of spermatogenesis, which itself has an absolute factor levels. While body and organ size in general are
requirement for androgen action. In both immature reduced in GH-deficient and IGF-1/2-deficient mice,
animals and those made experimentally gonadotropin spermatogenesis is complete in these small testes,
deficient, spermatogenesis can be established and suggesting no direct involvement of GH and growth
maintained by a direct action of testosterone on the factors in initiation of spermatogenesis (Sjogren et al.
seminiferous tubules (see Section 6.5). Rat models 1999).
have been used extensively to explore minimal and An interesting pathway for stimulating testoster-
optimal iTT levels required for the initiation, restor- one production by Leydig cells has apparently
ation or maintenance of spermatogenesis. Increases in developed in the common marmoset (Callithrix jac-
iTT can be induced by administration of exogenous chus). In the intact and normal marmoset, LH
testosterone in high dose (passive diffusion) or by receptor exon 10, although genomically present, is
hCG/LH administration (Leydig cell stimulation). not expressed (Zhang et al. 1997). For the human
An excellent model is the hypogonadal (hpg) mouse LH receptor, this exon is necessary for the expres-
that lacks endogenous gonadotropin secretion, in which sion of receptor protein (Zhang et al. 1998). Inter-
exogenous testosterone treatment induced spermato- estingly, a clinical case lacking LH receptor exon 10
genesis and sperm capable of in-vitro fertilization has been described (Gromoll et al. 2000). This boy
(Singh et al. 1995). had developed a male phenotype but presented with
In immature non-human primates, the adminis- retarded pubertal development, small testes and
tration of very high doses of testosterone will delayed bone maturation, all indicative of androgen
induce complete spermatogenesis, albeit to a limited deficiency. Given the similarity to marmoset LH
degree (Marshall et al. 1984). A remarkable clinical receptor status, this patient was successfully treated
corollary is the finding of spermatogenesis in with hCG, indicating that exon 10 is involved in
regions adjacent to Leydig cell tumors, presumably differential LH/hCG recognition. More recently it
due to locally high iTT (Weinbauer and Nieschlag was found that marmoset pituitary expresses hCG
1996). In boys with activating mutations of the LH (Gromoll et al. 2003), raising the possibility that
receptor, testosterone alone (in the absence of marmoset Leydig cells are driven by hCG rather
measurable FSH) can initiate precocious puberty than LH. It would be interesting to know the
and maturation of the testis (Shenker et al. 1993; dependence of marmoset spermatogenesis on iTT.
Gromoll et al. 1998). Conversely, patients with In any case, it is obvious that the control of Leydig
inactivating mutations of the LH receptor are both cell function and testosterone production by LH
poorly virilized and have small testes, suggesting an is entirely different in humans and marmosets
impairment of germ cell development in association (Michel et al. 2007).

128
Chapter 6: The role of testosterone in spermatogenesis

6.4.3 Adult spermatogenesis: testosterone and estradiol administration suppresses


LH secretion, iTT levels fall to 3% of normal, and
maintenance and reinitiation elongated spermatid production ceases (ODonnell
There are many animal, non-human and human et al. 1994; 1999). When larger doses of exogenous
model studies establishing that testosterone alone testosterone are given, elongated spermatid produc-
can maintain sperm production in adulthood. Testos- tion recommences; however, iTT concentrations do
terone, in the absence of FSH, can maintain at least not always show a significant rise (ODonnell et al.
qualitatively normal spermatogenesis in adult rats 1996a; 1999). Hence small changes in iTT levels
(Huang et al. 1987; Zirkin et al. 1989; Awoniyi et al. result in large changes in sperm output. In adult rats
1992; McLachlan et al. 1994a) and monkeys (Marshall with slightly suppressed FSH, spermatid production
et al. 1983; 1986) (see Section 6.6 for a discussion was suppressed when iTT was reduced to 4.24.6%
on the likely requirement for FSH in combination of normal, but was maintained when the testosterone
with testosterone to maintain quantitatively normal level was held at 7.48.8% of normal (Sun et al.
spermatogenesis). While LH is the primary driver 1990). The level of testosterone needed to maintain
of testosterone secretion, LH-independent production spermatogenesis is at least fourfold lower than the
of small amounts of androgen appears able to main- concentration present in the normal testis (Huang
tain low levels of sperm production in mice et al. 1987). An iTT level of approx 20 ng/ml
(Zhang et al. 2003). ( 70 nmol/l) is sufficient for the maintenance of
Suppression of LH/FSH secretion followed by complete spermatogenesis in rats (Awoniyi et al.
selective LH/testosterone replacement has been used 1989; Zirkin 1998). This threshold concentration of
to demonstrate that testosterone alone can maintain testosterone is still approximately four to eightfold
or restore qualitatively normal spermatogenesis. greater than serum testosterone levels, and greatly
Suppression of gonadotropins can be achieved by exceeds the dissociation constant of the testicular
GnRH immunization or antagonist administration AR (3 nmol/l). How the seminiferous epithelium
(Chandolia et al. 1991a; Awoniyi et al. 1992; responds to such small changes in androgen concen-
McLachlan et al. 1994b), removal of the pituitary trations in a setting where the AR is likely fully
(Huang et al. 1987) or by the administration of sex saturated is unclear. It is also intriguing to note that
steroids (testosterone  estradiol) (Awoniyi et al. the threshold dose of testosterone required to initiate
1989; Zirkin et al. 1989; McLachlan et al. 1994a; spermatogenesis in the mouse is about an order of
ODonnell et al. 1994). It is worthwhile noting that magnitude greater than that needed for the mainten-
sex steroid administration in rats causes marked ance of adult spermatogenesis, suggesting that two
decreases in LH with only minor changes in FSH, different mechanisms of androgen action may exist
but suppresses both LH and FSH in monkeys in the testis (Handelsman et al. 1999) depending on
and men. The subsequent restoration of iTT maturational status. Again, the mechanism of this
levels can be achieved by LH or hCG in primates, effect is unknown.
or in rodents by the administration of large doses In patients with hypogonadotropic hypogonad-
of exogenous androgen which diffuse into the ism, hCG (as an LH substitute) is used to stimulate
testis from the circulation (LH or hCG cannot be Leydig cell androgen secretion and elevate iTT levels
chronically administered to rodents due to antibody to initiate or restore spermatogenesis in pre- or post-
formation). This administration of high-dose testos- pubertal onset cases, respectively. In brief, in prepu-
terone in rodents likely has systemic effects, and in bertal cases the initiation of spermatogenesis
the case of the GnRH-immunized rodent, can adequate for fertility often requires the addition of
stimulate the pituitary to secrete FSH (McLachlan FSH (by pulsatile GnRH or administration of hMG
et al. 1994b), necessitating the use of alternate or urinary/recombinant human FSH) after several
strategies, such as immunoneutralization of FSH months of initial hCG therapy (Bchter et al.
(Meachem et al. 1998), to explore the specific role of 1998); while in men with adult onset hypogonado-
testosterone. tropic hypogonadism, hCG alone will often suffice
The maintenance or reinitiation of adult sperm- (Finkel et al. 1985; Burris et al. 1988; Hayes and
atogenesis by testosterone occurs over a narrow dose Pitteloud 2004). However, in patients with hypogo-
range (Zirkin 1998). For example in rats, low-dose nadotropic hypogonadism in whom spermatogenesis

129
Chapter 6: The role of testosterone in spermatogenesis

had been induced with hCG/hMG, when continuing 6.5 Androgen action on
on hCG alone to maintain normal peripheral testos-
terone levels, the sperm output persisted but waned spermatogenesis
over time (Depenbusch et al. 2002). Earlier reports 6.5.1 Testicular androgen production
also show that hCG maintained spermatogenesis in
men with hypogonadotropic hypogonadism (John- and metabolism
sen 1978; Vicari et al. 1992) but to a submaximal Testosterone is produced by the interstitial Leydig cells
extent. In support is the classic series of studies by under LH stimulation, resulting in iTT levels vastly
Matsumoto and Bremner, where hypogonadotropic greater than serum levels in all species examined. In
hypogonadism was experimentally induced in man they are 50200-fold higher but vary over a
normal men, and the ability of hCG and/or FSH to 510-fold range amongst healthy men (Morse et al.
maintain or restore spermatogenesis was examined 1973; Jarow et al. 2001; McLachlan et al. 2002a;
(Matsumoto et al. 1983; Matsumoto and Bremner Matthiesson et al. 2005). Recently Roth et al. (2010a)
1985; Matsumoto et al. 1986). These studies con- confirmed the relationship between serum LH
cluded that while qualitatively normal spermatogenesis and iTT.
was maintained by either hormone, quantitatively The concentration gradient between the testis
normal sperm output required both gonadotropins. and circulation is maintained even when Leydig cell
Thus, maintenance and reinitiation of spermato- steroidogenesis is experimentally suppressed (Zirkin
genesis by testosterone/hCG in patients with hypogo- et al. 1989). In rodents, it has been suggested that the
nadotropic hypogonadism is possible (Nieschlag et al. testis acts as a reservoir through testosterone bind-
1999) with some difference in outcome likely to result ing with high affinity to androgen binding protein
from patient differences variable etiology, time of (ABP), a Sertoli cell product under androgen and
onset (pre- and post-pubertal), preceding therapy, FSH control (Jarow and Zirkin 2005). Transgenic
prior history of cryptorchidism and pre-therapy over-expression of testicular ABP, which could theor-
testicular volume (Liu et al. 2002; 2009). Comparing etically reduce testosterone bioavailability, causes
doses, the reinitiation of spermatogenesis seems to germ cell apoptosis (Jeyaraj et al. 2003). However,
require more testosterone either higher doses or the molar concentrations of iTT far exceed that of
longer duration of exposure than does the mainten- ABP, and thus the extent to which ABP modulates
ance of spermatogenesis. This became particularly testosterone bioavailability and action is unclear. Also
evident from studies using GnRH analog treatment and its relevance to the human is moot as ABP is not
testosterone substitution in a hormonal contraceptive made within the testis, with the sex-steroid binding
context; wherein concomitant testosterone supple- protein, SHBG, being produced by the liver and
mentation prevented the induction of azoospermia, entering the circulation.
but delayed substitution with the same dose of testos- In man the alternative possibility for the con-
terone failed to do so (Weinbauer and Nieschlag 1996). tinued excess of iTT relative to serum testosterone
In summary, data from animal and human models in the presence of immeasurably low serum LH
in a wide variety of settings support the following (<0.5% baseline using supersensitive methods
concepts: (Robertson et al. 2001)) is the existence of LH-
 In adulthood iTT levels are 50100-fold higher independent constitutive androgen secretion. While
than serum levels. low iTT levels ( 23% baseline) are often associated
 Intratesticular testosterone levels at 520% of with a complete failure of spermatogenesis, in some
normal can support spermatogenesis, and even settings they may support a limited degree of sperm-
low levels (e.g. 2% of normal adult levels) can atogenesis. This provides a potential explanation
support a degree of spermatogenesis in some for the failure of androgen-based male hormonal
species/individuals. contraceptive strategies (that rely exclusively on gona-
 A narrow dose-response range exists between iTT dotropin withdrawal) to be universally effective, with
levels and sperm output; small changes in iTT 5% of normal men maintaining sperm densities
concentrations can produce large changes in sperm >1 million/ml (Liu et al. 2008). Establishing the
output (see Section 6.5.4 for further discussion). absence of androgen action in any model is essentially
impossible due to constraints of assay methods.

130
Chapter 6: The role of testosterone in spermatogenesis

Yet several indirect lines of evidence for continued with LH suppression, while DHEA less so; the 50%
androgen action following shutdown of the HPT fall in serum androstenedione shows the testis to be a
axis can be found in both animals and man. For major source of this steroid, but unchanged serum
example, a further deterioration in spermatogenesis DHEA suggests this is predominantly of adrenal
is seen with the addition of the androgen receptor origin (Roth et al. 2011).
inhibitor, flutamide, to GnRH antagonist-treated Testosterone is metabolized by the 5a-reductase
rats (Kangasniemi et al. 1996). This concept provides enzyme to DHT which is a potent androgen. Dihy-
a ready explanation for the eventual establishment drotestosterone is further metabolized in the testis by
of spermatogenesis in the LH receptor knock-out 3a-hydroxysteroid dehydrogenase to 5a-androstane-
mouse (Zhang et al. 2003). In a human androgen 3a,17b-diol (3a-adiol). This metabolite interacts only
progestin contraceptive context, support comes from weakly with the AR; however, it can be readily con-
the finding of significant (1355 nM) iTT levels in verted back to DHT and thus can act as an androgenic
men with spermatogenic suppression (Jarow et al. precursor, at least in some species (Wilson et al.
2001; McLachlan et al. 2002a), and evidence that high 2003). In many peripheral tissues where testosterone
doses of exogenous testosterone may allow sufficient levels are comparatively low, the 5a-reduction of tes-
back diffusion to reduce contraceptive efficacy tosterone to the more potent androgen DHT is neces-
(Meriggiola et al. 2002; Liu et al. 2008). Finally, data sary for the full expression of androgen action
suggest an improved contraceptive effect with the (Deslypere et al. 1992). This is because, at equimolar
inclusion of CPA, a progestin with AR-inhibiting concentrations, DHT binds with higher affinity to,
activity, in a hormonal contraceptive regimen (Mer- and forms a more stable complex with, the AR than
iggiola et al. 1998). The use of high-dose CPA in men testosterone, and is consequently much more potent
with prostate cancer has been reported to severely in inducing an androgenic response in various assays
inhibit spermatogenesis (Re et al. 1979), an effect that and reporter systems (Grino et al. 1990; Deslypere
one could speculate relates to more complete loss of et al. 1992; Zhou et al. 1995; Jarow and Zirkin
androgen action that goes above and beyond that due 2005). In the normal testis DHT levels are much lower
to gonadotropin withdrawal. than testosterone on a molar basis and thus unlikely
While one can hypothesize that the complete to be a player in androgenic stimulation of sperm-
elimination of androgen action from the human atogenesis under normal settings, despite its higher
seminiferous epithelium would ensure universal potency.
azoospermia, it is a difficult notion to pursue as it However, the 5a-reduction of testosterone may
must specifically target the testis whilst maintaining be important for androgen action on spermatogen-
non-gonadal actions essential for physical and psycho- esis in situations where iTT levels are low. During
logical health. Potential strategies might be to target the pubertal initiation of spermatogenesis in
testis androgen synthesis and/or AR-responsivity, rodents, DHT and 3a-adiol are the predominant
for example an inhibitor of the 17b-hydroxysteroid androgens present in the testis (Corpechot et al.
dehydrogenase type 3, or alternatively, a testis-specific 1981), and there is a peak in the testicular activity
inhibitor of the AR or its coactivators. A much better of both 5a-reductase isoenzymes (Killian et al.
understanding of testosteroneAR signaling in the 2003). Investigation of whether 5a-reduction to
testis is clearly needed. DHT is required during the initiation of spermato-
Testosterone is the major androgen found within genesis is hampered by the fact that inhibition of
the testis, with levels far exceeding that of other 5a-reductase causes an increase in testosterone to
metabolites (Jarow and Zirkin 2005; ODonnell levels that are likely able to act directly on the AR
et al. 2006). Testicular testosterone is synthesized without amplification (George et al. 1989; Killian
from precursors, androstenedione and DHEA. et al. 2003). Nevertheless, it seems likely that the
A recent study using testicular fluid aspiration in 5a-reduction of testosterone is important for the
normal and GnRH antagonist-treated normal men ability of testosterone to initiate spermatogenesis
has shown that their baseline concentrations are during puberty (ODonnell et al. 2006).
about 25% that of testosterone, implying rapid con- In adult rodents, 5a-reduction to DHT is import-
version to testosterone, and vastly in excess of their ant for spermatogenesis when iTT levels are experi-
serum levels. Androstenedione levels fell markedly mentally lowered, since the androgen-dependent

131
Chapter 6: The role of testosterone in spermatogenesis

restoration of germ cell numbers by a suboptimal dose of estradiol via a membrane-bound receptor (Carreau
of testosterone was impaired by the co-administration and Hess 2010; Carreau et al. 2011). The role of
of a 5a-reductase inhibitor (ODonnell et al. 1996a; testosterone metabolism to estradiol in spermato-
1999). This data, along with observations in men genesis is discussed in Section 6.6.3.
(Anderson et al. 1996), point to a potential role for
5a-reductase in maintaining androgen action during
male hormonal contraception when iTT levels are 6.5.2 Testicular androgen receptor
decreased. This concept has proved difficult to explore
in clinical studies, and to date no additive effects and sites of androgen action
have been seen from the inclusion of 5a-reductase The AR is expressed in Sertoli cells, Leydig cells and
inhibitors in a small number of men undergoing peritubular myoid cells (Bremner et al. 1994; Van
hormonal contraception (McLachlan et al. 2000; Roijen et al. 1995). The general consensus is that germ
Kinniburgh et al. 2001; Matthiesson et al. 2005). cells lack AR, despite some conflicting immunohisto-
In summary, testosterone is the major androgen chemical studies, reviewed in ODonnell et al. (2006).
acting on the AR during spermatogenesis, but the In any case, it seems that AR in germ cells is not
5a-reduction of testosterone to DHT may contribute essential for their development, since germ cells with-
at lower testosterone concentrations. out AR develop normally in testes which contain
Within the testis, testosterone is aromatized to functional AR (Johnston et al. 2001; Tsai et al.
estradiol catalyzed by the aromatase cytochrome 2006). Sertoli cells in the neonatal period show weak
P450 enzyme (P450arom, the product of the CYP19 AR immunoreactivity which progressively increases
gene). The testis is the major site of estrogen after day 14, when Sertoli cells cease proliferating and
production in the male, and prodigious levels of commence terminal differentiation, to reach adult
estradiol are present within testicular fluids and much levels at day 45. In the adult, the expression of the
higher than in the circulation in many species (Hess AR varies in relation to the stage of spermatogenesis.
2003). In normal men, the concentration of testicular The mid-spermatogenic stages (VIIVIII in the rat
estradiol is surprisingly high, with levels of 11 000 and mouse, and IIIII in the human) are considered
58 000 pmol/l (Zhao et al. 2004; Matthiesson et al. the most androgen responsive (Walker and Cheng
2005), and the testis : serum estradiol concentration 2005; ODonnell et al. 2006). Consistent with these
gradient (407-fold) is even larger than that of testos- findings, immunohistochemical studies show that AR
terone (120-fold) (Matthiesson et al. 2005). The aro- protein is maximal in these stages in rats (Bremner
matase enzyme is expressed in various testicular cell et al. 1994), monkeys (McKinnell et al. 2001) and man
types including Sertoli cells; while germ cells them- (Suarez-Quian et al. 1999).
selves are an important source of estrogen (Hess 2003; In recent years, data from transgenic mouse
Carreau and Hess 2010; Carreau et al. 2011). Estrogen models have provided important information on the
receptors, like ARs, are members of the steroid sites of testosterone and androgen action in sperm-
hormone family of receptors, and bind to estrogen atogenesis, reviewed in Wang et al. (2009). Ablation
response elements (EREs) in the promoter region of of AR in all tissues results in an androgen insensitivity
estrogen-responsive genes to modulate transcription. syndrome and failure of the urogenital tract to
The ER subtypes form homo- and heterodimers, and develop normally (Wilson 1992), making it difficult
the relative expression of each subtype within a par- to study the role of the AR in the initiation of sperm-
ticular cell determines the cells ability to respond to atogenesis. The dependence of spermatogenesis on
particular ligands (Hall and McDonnell 1999). Both androgen was highlighted by studies in which andro-
ER genes (ERa and ERb) are widely expressed in the gen (both testosterone and DHT), in the absence of
testis, with conflicting immunohistochemical data FSH, was able to initiate spermatogenesis in mice
reported for various species (reviewed in ODonnell congenitally deficient in GnRH (hpg mice) (Singh
et al. 2001b; Carreau and Hess 2010). The general et al. 1995). Using a Cre-lox conditional knock-out
consensus is that ERa predominates in Leydig cells strategy, two mouse lines were created in which func-
and ERb in the seminiferous epithelium, as reviewed tional AR was selectively ablated from Sertoli cells
in Carreau and Hess (2010). Germ cells are also (Chang et al. 2004; De Gendt et al. 2004). The result-
capable of responding to rapid, non-genomic actions ant spermatogenic arrest in Sertoli cell androgen

132
Chapter 6: The role of testosterone in spermatogenesis

Fig. 6.3 Diagram of sites of androgen


action in the testis. Luteinizing hormone
from the circulation stimulates Leydig
Elongated
cells to synthesize and secrete
spermatids
testosterone (T). Testosterone acts on
androgen receptors (ARs) within testicular
somatic cells. Testosterone can be further
metabolized to DHT, which also acts via
the ARs in a setting of reduced iTT (not
Round spermatid shown), primarily when testosterone
levels are low (see Section 6.5.1).
Androgens act on nuclear ARs in Sertoli
cells to modulate their function.
Spermatocyte Unknown signals (solid arrows) from the
Sertoli cells impact on androgen-
dependent germ cell processes,
particularly spermatocyte survival and
Spermatogonium progression through meiotic division, as
well as progression through
spermiogenesis. Unknown androgen-
regulated signals also modulate the
ability of the Sertoli cell to release
Sertoli cell elongated spermatids at spermiation.
Androgen also acts on ARs in peritubular
myoid cells (dashed arrow) to modulate
Peritubular Sertoli cell functions and, in turn, support
germ cell development though unknown
myoid cell
mechanisms. An autocrine action of
androgen on AR in Leydig cells is
T required for optimal steroidogenesis,
AR which in turn is important for
T
quantitatively normal spermatogenesis.
T

Leydig cells

LH

receptor knock-out (SCARKO) mice proved that the are mediated by AR in peritubular myoid cells (Welsh
initiation of spermatogenesis required androgen et al. 2009). The ablation of AR from many, but
action on the Sertoli cell. The ability of androgen to not all, peritubular cells results in azoospermia and
stimulate spermatogenesis involves a direct action infertility, and suggests that androgens act on AR
on Sertoli cell AR, because the androgen-mediated within peritubular cells which in turn modulate
induction of spermatogenesis in hpg mice requires Sertoli cell function and their ability to support
the presence of AR in Sertoli cells (OShaughnessy spermatogenesis (Welsh et al. 2009). Within Leydig
et al. 2010a). The effects of androgen on spermato- cells, AR is also required for optimal steroidogenesis
genesis rely on the classic, genomic AR pathway within to produce intratesticular androgens necessary for
Sertoli cells, because Sertoli cell expression of a mutant germ cell development (Tsai et al. 2006). Therefore
AR protein, which is unable to bind to androgen androgen action on AR within the somatic cells of the
response elements in DNA, causes spermatogenic testis is essential for spermatogenesis (Fig. 6.3).
arrest (Lim et al. 2009). Interestingly, however, at least In terms of androgen regulation of germ cell
some of the effects of androgen on spermatogenesis development, the initiation and maintenance of

133
Chapter 6: The role of testosterone in spermatogenesis

meiosis is well known to be particularly androgen to be sensitive to androgen withdrawal, with various
sensitive (Handelsman et al. 1999). In the absence of suppression regimes resulting in the failure of Ser-
androgen action, spermatogonia can enter meiosis; toli cells to release spermatids (ODonnell et al.
however, spermatocyte survival during the long mei- 2011). Spermatids that are not released are instead
otic prophase, and their subsequent meiotic division, rapidly phagocytosed by the Sertoli cell. Spermia-
relies on androgen action on Sertoli cell AR. This tion failure is an acute response to androgen and
is evidenced by the phenotype of SCARKO mice, gonadotropin suppression (Russell and Clermont
which displayed meiotic arrest, with very few haploid 1977), and sperm release from the testis is reduced
spermatids produced (Chang et al. 2004; De Gendt by 5090% within the first week (Saito et al. 2000;
et al. 2004). Although spermatogonial numbers were ODonnell et al. 2009). Spermiation failure is a
normal, SCARKO mice displayed elevated apoptosis major factor in the rapid (within four to six weeks)
and a progressive loss of pachytene spermatocytes suppression of sperm counts seen in men with
after stage VII, indicating that testosterone acts via reduced intratesticular androgen levels resulting
the Sertoli cell AR to support spermatocyte survival from androgen-based hormonal contraception
(De Gendt et al. 2004). The final meiotic division was (McLachlan et al. 2002a; Matthiesson et al. 2006).
also markedly compromised, with observations sug- Androgen suppression alone can cause spermiation
gesting a failure of diplotene spermatocytes to enter failure (Saito et al. 2000), and we have noted that
the first meiotic division (Chang et al. 2004). These the addition of an AR antagonist to an androgen
studies demonstrate that the completion of meiosis is suppression regime results in more profound sper-
absolutely dependent on androgen action via the Ser- miation failure (Saito et al. 2000; ODonnell et al.
toli cell AR; however, the precise androgen-dependent 2009). Observations in mice expressing AR with
signals from the Sertoli cells and how they regulate reduced function highlight that spermiation is sen-
survival and division of spermatocytes remain sitive to reduced AR signaling (Holdcraft and Braun
unknown. 2004a). Sperm release in vitro can be reduced by the
Androgen action is also well known to be required administration of an AR antagonist (Shupe et al.
for the initiation, maintenance and restoration of 2011). However, this process is unlikely to rely solely
haploid germ cell development (spermiogenesis; upon androgens, as various other endocrine disturb-
reviewed in ODonnell et al. 2006; Ruwanpura et al. ances also induce acute spermiation failure (reviewed
2010). In adult rats, the suppression of iTT to 3% of in ODonnell et al. 2011), and numerous kinase and
normal causes the premature detachment of round phosphatase modulators can modulate sperm release
spermatids from Sertoli cells at the beginning of the in vitro (Chapin et al. 2001).
elongation phase, thereby preventing the production It is generally considered that androgens are not
of elongating and elongated spermatids (ODonnell required for the development of spermatogonial
et al. 1996b). This premature detachment can be populations in rodents (e.g. Haywood et al. 2003;
completely restored within four days of testosterone De Gendt et al. 2004). Spermatogonia are highly
replacement (ODonnell et al. 1994). These observa- responsive to changes in FSH (Haywood et al.
tions, along with others (Cameron and Muffly 1991), 2003); thus the decreased numbers seen in gonado-
suggest that androgens act to modulate intercellular tropin-suppressed in men are likely a consequence of
adhesion between Sertoli cells and spermatids, specif- reduced FSH, rather than androgenic support
ically at a time when the spermatid forms specialized (McLachlan et al. 2002a; Ruwanpura et al. 2010).
adhesion junctions with the Sertoli cell (McLachlan However, androgens may be able to maintain
et al. 2002b). Androgen also maintains round sperm- adult spermatogonial populations in men when
atid survival, particularly in stage VII and VIII FSH levels are reduced, at least in the short term
(ODonnell et al. 2006). Spermiogenic arrest is also (Matthiesson et al. 2006). Although direct action on
seen in various transgenic models where androgen the Sertoli cell AR does not seem to be required for
action is reduced (e.g. Holdcraft and Braun 2004a; spermatogonial development (De Gendt et al. 2004),
Pakarainen et al. 2005; Lim et al. 2009). spermatogonia were reduced in mice with total tes-
The final release of sperm from the Sertoli cell at ticular AR ablation (Tan et al. 2005). This latter
the end of spermatogenesis occurs via a process observation may be a consequence of the abdominal
known as spermiation. Spermiation is well known location of the testes in these mice, leading to

134
Chapter 6: The role of testosterone in spermatogenesis

compromised Sertoli cell function, or may reflect a and hence create a permeability barrier and to main-
role for androgen support of spermatogonia outside tain testicular immune privilege has recently been
of the seminiferous epithelium, such as via peritub- shown to be androgen dependent (Meng et al.
ular myoid cells (Tan et al. 2005). Consistent with 2005; Kaituu-Lino et al. 2007; Meng et al. 2011).
this, ablation of AR from most, but not all, peritub- Finally it is worth noting that studies in non-
ular cells (PTM-ARKO mice) leads to a reduced human primates revealed that testosterone induced
number of spermatogonia in adult mice (Welsh the appearance of a-smooth muscle actin in the peri-
et al. 2009). This may reflect an androgen-mediated tubular cells of the testis (Schlatt et al. 1993). These
effect on peritubular myoid cell secretion of growth results are supported by observations in peritubular
factors and/or basement membrane deposition, cells which lack AR in a transgenic mouse line (Welsh
which may in turn influence spermatogonial devel- et al. 2009). These observations suggest that testoster-
opment (Welsh et al. 2009). However, high iTT one initiates the contractile function of these cells,
levels were observed in PTM-ARKO mice, which which may be important for movement of sperm
may inhibit spermatogonial development. An inhibi- along the seminiferous tubules. Recent studies in mice
tory effect of high serum/intratesticular testosterone in which AR was ablated from testicular arteriole
on spermatogonia has been observed in adult (SMC) suggest a role for androgens in the control of
rodents (Meachem et al. 1998; Shetty et al. 2001), vasomotion and fluid dynamics in the testis (Welsh
but whether this phenomenon occurs in primates is et al. 2010).
unclear.
As evidenced throughout this section, many 6.5.3 Mechanisms of androgen action
effects of androgens on spermatogenesis are medi-
ated via Sertoli cells. Androgens, acting on the AR, in the testis
modulate the expression of various genes and pro- Androgens act on intracellular receptors and can acti-
teins within Sertoli cells (De Gendt et al. 2004; vate different pathways to modulate cellular function
Tan et al. 2005), which in turn mediate the Sertoli during spermatogenesis (Walker 2009). The classic
cells ability to support germ cell development; see or genomic pathway of AR action involves androgen
Section 6.5.3. Observations in transgenic mice indi- (testosterone or DHT) diffusing through the plasma
cate that neonatal Sertoli cell proliferation, and hence membrane and interacting with AR which have
the attainment of a normal Sertoli cell population, is previously been bound to heat shock proteins in the
unaffected by an absence of AR signaling via the cytoplasm. Androgen binding to the AR causes a
Sertoli cell (De Gendt et al. 2004; Tan et al. 2005; conformational change, or activation, of the ligand-
Lim et al. 2009), but is dependent on FSH action receptor complex such that it attains high affinity for
(Allan et al. 2004). However, androgen action via the specific binding sites within DNA. The activated
genomic AR pathway outside the Sertoli cell may be receptor translocates to the nucleus and binds to
important for the FSH-mediated effects on Sertoli sequences, known as androgen response elements
cell populations (Schauwaers et al. 2007; Lim et al. (AREs), in the promoter region of androgen-responsive
2008; 2009); a mechanism which may explain the genes. The DNA binding initiates the recruitment of
reduced Sertoli cell numbers in certain models with coactivators or co-repressor proteins that modulate
ablation of AR from all testicular cells, but no reduc- gene transcription. This genomic action of AR causes
tion when AR is ablated from Sertoli cells only changes in gene transcription in 3040 min, and
(De Gendt et al. 2004; Johnston et al. 2004; Tan changes to cellular proteins several hours later
et al. 2005). This mechanism may be mediated in (Walker 2009). Androgen response elements can be
part by androgens acting on peritubular myoid cells non-selective, in that they can respond to progester-
to modulate paracrine factor secretion and/or base- one or glucocorticoid receptors, or selective, in that
ment membrane deposition important for Sertoli cell they respond to AR only. That selective vs. non-select-
proliferation (Welsh et al. 2009). It is clear from ive AREs drive different biological processes in
many studies that androgens are essential for the spermatogenesis was recently demonstrated by the
differentiation and maturation of Sertoli cells and finding of reduced germ cell numbers in the male
for their ability to support germ cell development. SPARKI (Specificity-Affecting AR knockIn) mouse
The ability of Sertoli cells to form tight junctions (Schauwaers et al. 2007). In this model, the second

135
Chapter 6: The role of testosterone in spermatogenesis

zinc finger of the AR DNA-binding domain was et al. 2009). The resulting phenotype recapitulated
replaced with that of the glucocorticoid receptor, the phenotype of SCARKO mice (i.e. predominant
resulting in an AR that binds non-selective AREs meiosis defect), suggesting that genomic AR mech-
but is unable to recognize selective AREs. Spermato- anisms are essential for the androgen-mediated initi-
genesis was compromised in these mice, but the ation of spermatogenesis, and providing indirect
spermatogenic phenotype differed from SCARKO evidence that non-genomic AR pathways play no
mice, in which both selective and non-selective ARE major role (Lim et al. 2009). However, the gener-
responses are impaired. Therefore studies in the ation of transgenic mouse models or pharmaco-
SPARKI mice show that androgen responses during logical strategies in which the non-genomic AR
spermatogenesis are mediated by both selective and pathway is specifically affected in vivo is needed to
non-selective AREs. elucidate the role of non-genomic androgen actions
At least two non-genomic pathways of androgen in spermatogenesis.
action have been proposed to participate in the The precise genes and proteins regulated by
response of Sertoli cells to androgen stimulation androgens in Sertoli cells, and other testicular cells,
(reviewed in Walker 2010). One involves the rapid are not well understood. The best characterized
(2040 s) and transient influx of calcium into Ser- androgen-responsive gene in Sertoli cells is Rhox5,
toli cells, which appears to involve K(+)ATP chan- formerly known as Pem (Lindsey and Wilkinson
nels, activation of an unknown G-protein-coupled 1996), which may act as a key intermediate in
receptor and the phospholipase C pathway (Loss modulating androgen action on Sertoli cells (Hu
et al. 2004), reviewed by Walker (2010). The second et al. 2010). Other androgen-induced genes have
is an AR-dependent pathway involving ligand- been discovered in the SCARKO model, including
bound AR in the plasma membrane. Although stud- Tubb3 and Spinlw (Eppin) (OShaughnessy et al.
ies suggest that a small population of ARs exists 2010b). While some genes are directly modulated
near the plasma membrane, there is as yet no by an ARE-dependent mechanism, many other
definitive evidence of AR within the membrane genes are modulated indirectly, via androgen-
(Walker 2010), and no immunohistochemical evi- induced changes in transcription or paracrine
dence of membrane AR localization in the semin- factors (OShaughnessy et al. 2010b). Microarray
iferous epithelium. In this model of non-genomic analysis of androgen-dependent seminiferous
AR action, ligand-bound AR recruits and activates tubules undergoing acute androgen and FSH sup-
Src kinase, which in turn activates the EGF recep- pression revealed transcriptional changes in both
tor. These events activate the ERK-MAP-kinase Sertoli cells and germ cells, with cohorts of genes
pathway and ultimately, phosphorylation of the being up- or downregulated in a cell- and stage-
transcription factor CREB. Phosphorylated CREB specific manner (ODonnell et al. 2009), supporting
then modulates the transcription of CREB-regulated the concept that androgen action in the testis regu-
genes (reviewed by Walker 2010). lates a cascade of transcriptional changes within the
Investigation into the different androgen-depend- seminiferous epithelium.
ent molecular pathways and their specific roles in We recently demonstrated that androgens can
spermatogenesis is hampered by the fact that such modulate miRNAs within Sertoli cells (Fig. 6.4), and
studies largely rely on observations in isolated Sertoli that these miRNAs in turn influence protein transla-
cells. As such it has been difficult to determine the tion (Nicholls et al. 2011). We have also observed
relative contributions of the genomic and non-geno- changes in germ cell proteins in response to androgen
mic pathways to androgen-dependent processes such suppression and replacement (ODonnell and Stan-
as meiosis and spermiogenesis. In-vitro studies sug- ton, unpublished data). Taken together, androgen
gest that the non-genomic AR pathway may contrib- action in vivo mediates spermatogenesis by modulat-
ute to the regulation of Sertoli cellgerm cell ing, directly and indirectly, gene and protein expres-
adhesion and spermiation (Shupe et al. 2011). Geno- sion within Sertoli cells and germ cells. How the germ
mic AR actions were assessed by the selective disrup- cells respond to this androgenic stimulus is unclear,
tion of the AR DNA-binding domain in Sertoli cells, but likely involves androgen-induced changes in cell
thus theoretically eliminating genomic AR mechan- surface receptors or paracrine factor production by
isms but preserving non-genomic mechanisms (Lim Sertoli cells.

136
Chapter 6: The role of testosterone in spermatogenesis

doses (ODonnell et al. 1996a); however, there is


currently no explanation for why small changes in
Protein testicular androgen concentration have dramatic
+
consequences for spermatogenesis. What is certain,
miR
though, is that different processes in spermatogenesis
have different sensitivities to, or requirements for,
1.5 androgens. This concept has been demonstrated
consistently in transgenic mouse models and in rat
suppression and replacement models.
T
When promoting spermatogenesis in the congeni-
T
tally GnRH-deficient hpg mouse, the survival of
T+ flut spermatocytes and their entry into meiotic division
1.0
was stimulated by comparatively low doses of andro-
gens that are unable to restore spermatid elongation
miR 690

(Singh et al. 1995; Handelsman et al. 1999). Higher


levels of androgen will promote spermatid elongation
(Handelsman et al. 1999), probably via the androgen-
0.5 dependent effects on mid-spermiogenesis (ODonnell
et al. 1994). These observations are supported by the
phenotypes of transgenic mouse lines with variable
inactivation of AR action within Sertoli cells. Ablation
of AR function in Sertoli cells causes meiotic arrest
0.0 and very limited spermiogenesis (Chang et al. 2004;
0 3 6 De Gendt et al. 2004; Lim et al. 2009); whereas
Time (h) reduced (but not ablated) androgen signaling or
Fig. 6.4 Androgen effects on miRNAs in Sertoli cells. Sertoli cells production can support meiosis but spermiogenesis
were isolated from 20-day-old rats and cultured in the absence of is disrupted (Zhang et al. 2001; Holdcraft and Braun
hormones for four days, prior to the addition of FSH and 2004a; Lim et al. 2009). Thus the completion of mei-
testosterone (T), with or without the androgen receptor antagonist
flutamide (flut) for 06 hours. Levels of one miRNA species (miR 690) osis requires less androgen action on the Sertoli cell
were measured by quantitative PCR and normalized to GAPDH than does the completion of spermiogenesis. In our
(glyceraldehyde 3-phosphate dehydrogenase). Data is shown as rat studies, levels of testosterone around 3% of normal
mean of three separate experiments, adapted from (Nicholls et al.
2011). In this instance, T treatment rapidly suppresses this miRNA, will maintain meiosis; whereas levels 12% of normal
and the T-mediated inhibition is blocked by the AR antagonist. Since are required for elongated spermatid production
miRNAs typically act by suppressing translation of their target (ODonnell et al. 1994; 1999).
protein, this T-mediated inhibition of miR 690 would be expected to
result in increased translation of its target protein(s). Acute There is also a dose effect of androgen on different
regulation of miRNA species in Sertoli cells may be one mechanism processes within spermiogenesis, at least in rodents.
by which androgens can modulate cellular responses. When testosterone levels are reduced to about 3% of
normal, the survival of step 7 round spermatids is
maintained, but the adhesion of step 8 round sperm-
6.5.4 Relationship between testicular atids to Sertoli cells is not supported; further with-
androgen concentrations and drawal of androgenic support, by an AR antagonist or
5a-reductase inhibitor, induces apoptosis of step 7
spermatogenic progression round spermatids (ODonnell et al. 1996a; 1999).
As discussed in Section 6.4.3, spermatogenesis can be The release of sperm from Sertoli cells at the end of
supported by levels of androgen that are about one- spermiogenesis also requires considerable androgenic
quarter of the levels present in the normal adult testis, support, and we have noted that levels of testosterone
and a very narrow dose-response range exists in (8% of normal in adult rats) that support elongated
rodents between testicular androgens and sperm pro- spermatid production at 50% of normal do not fully
duction. This latter observation may in part reflect the support spermiation, with many spermatids being
involvement of 5a-reduction of testosterone at lower retained by Sertoli cells, and thus reducing the sperm

137
Chapter 6: The role of testosterone in spermatogenesis

output from the testis (ODonnell et al. 1999 and testicular androgens and sperm counts (Weinbauer
unpublished data). et al. 2001; Narula et al. 2002). Because of the ability
Inhibition of androgen action will first result in of low levels of androgen action to support some
the failure of spermatid release (Russell and Clermont aspects of spermatogenesis, it is likely that novel
1977; Saito et al. 2000), followed by a more gradual approaches will be needed to diminish intratesticular
decline in germ cell populations (Saito et al. 2000). androgen action to the extent required for complete
This indicates that spermiation in the adult is acutely spermatogenic suppression in a contraceptive setting.
sensitive to a loss of androgen (and/or gonadotropin)
action (reviewed in ODonnell et al. 2011). This con- 6.6 Other endocrine factors
cept explains why spermiation failure is seen in
men undergoing acute gonadotropin withdrawal in the control of spermatogenesis
(McLachlan et al. 2002a). In these men, LH and FSH 6.6.1 Follicle-stimulating hormone
are undetectable by highly sensitive assays (Robertson
et al. 2001; McLachlan et al. 2002a), and iTT and and spermatogenesis
DHT are reduced to 2.5% and 65% of normal, The specific roles for FSH in spermatogenesis have
respectively. The maintenance of some testicular been reviewed extensively elsewhere (Nieschlag et al.
androgen action in these men likely explains why 1999; Holdcraft and Braun 2004b; ODonnell et al.
their germ cells progressed through meiosis and sper- 2006; OShaughnessy et al. 2009; Ruwanpura et al.
miogenesis (McLachlan et al. 2002a); however, the 2010) and therefore will only briefly be considered
number of cells entering meiosis was reduced as a here. Follicle-stimulating hormone receptors are pre-
consequence of FSH suppression resulting in lower sent only on Sertoli cells (Heckert and Griswold
numbers of meiotic precursor cells (spermatogonia) 1993), reflecting the importance of this gonadotropin
(see Section 6.6.2). for Sertoli cell function. Testicular FSH receptor
In summary, it is clear that some aspects expression in the rat is highest in stages XIIIII
of spermatogenesis, particularly the completion of (Heckert and Griswold 1993; Rannikko et al. 1996);
meiosis, can be maintained by very low levels of e.g. when type A spermatogonia proliferate and the
androgen action in the testis, and that different meiotic division is being completed. In the human
androgen-dependent processes within spermatogen- testis, FSH receptor expression could not be clearly
esis have different thresholds for androgen action. associated with a particular spermatogenic stage(s) but
There may be considerable between-subject variation was unequivocally confined to Sertoli cells (Bckers
in the specific androgenic requirement for each et al. 1994). There has been considerable confusion in
androgen-dependent process, as is suggested by stud- the literature over the specific roles of FSH, independ-
ies in humans (Zhengwei et al. 1998; McLachlan et al. ent of androgens, in spermatogenesis. This is in part
2002a; Matthiesson et al. 2005). Spermiation failure, probably due to the fact that FSH can support a low
which has a major influence on sperm output from level of LH-independent androgen production by
the testis, also shows a highly variable response to Leydig cells (e.g. Haywood et al. 2002; 2003; reviewed
androgen suppression in monkeys (ODonnell et al. in OShaughnessy et al. 2009), making it more diffi-
2001a), men (Matthiesson et al. 2005) and even in cult to study the effects of FSH in a complete absence
rodents (Saito et al. 2000; ODonnell et al. 2009). Thus of androgen action.
individual variations in the sensitivities of different Follicle-stimulating hormone has a key role in
spermatogenic processes to androgens may explain regulating the proliferation and function of Sertoli
why a correlation between iTT levels and sperm cells (Means et al. 1980; Wreford et al. 2001; Allan
output has been so difficult to establish in gonado- et al. 2004; Grover et al. 2004; Abel et al. 2008;
tropin-suppressed monkeys and men (Zhengwei et al. reviewed in OShaughnessy et al. 2009; Ruwanpura
1998; Weinbauer et al. 2001; Narula et al. 2002; et al. 2010). Follicle-stimulating hormone regulates
Matthiesson et al. 2006). In these models, the extent postnatal, but not fetal, Sertoli cell proliferation to
of FSH suppression in response to exogenous andro- establish a quantitatively normal, functional popula-
gen administration and the ability to support sperma- tion of Sertoli cells. Given that the number of Sertoli
togonial populations (see Section 6.6) likely adds cells is a key determinant of adult sperm output, this
further complexity to the correlation between function of FSH is important for quantitatively

138
Chapter 6: The role of testosterone in spermatogenesis

normal spermatogenesis. This is evidenced by trans- monkeys to achieve azoospermia as opposed to oli-
genic mouse models lacking FSH action that show gospermia was associated with lower FSH levels
reduced sperm counts due to a smaller Sertoli cell which in turn likely resulted in lower numbers of
population and various defects in the ability of Sertoli B spermatogonia (ODonnell et al. 2001a). Therefore
cells to support germ cell development (Wreford the degree of FSH suppression, as well as androgen
et al. 2001; Allan et al. 2004; Grover et al. 2004). suppression, is likely important for the induction of
Follicle-stimulating hormone also has a key role in azoospermia in a gonadotropin-suppressed, contra-
spermatogonial development as evidenced by studies ceptive setting. Follicle-stimulating hormone alone
in rodents (Haywood et al. 2003; Abel et al. 2008) and can maintain human germ cell development for six
primates (Weinbauer et al. 1991; Marshall et al. 1995). weeks during gonadotropin suppression, further
Follicle-stimulating hormone regulates spermatogo- emphasizing FSHs permissive role in human sperm-
nial populations by preventing apoptosis and possibly atogenesis, and the fact that optimal contraceptive
by supporting proliferation (reviewed in Ruwanpura suppression requires suppression of both LH and
et al. 2010). In monkeys and men, type A and B FSH (Matthiesson et al. 2006). In considering thresh-
spermatogonia are highly sensitive to gonadotropin old serum levels of FSH, or LH, action on human
withdrawal; although there are differences between spermatogenesis, highly sensitive assay methods
the sensitivity of spermatogonial subtypes depending should be used as these may reveal interesting rela-
on the experimental context. Follicle-stimulating tionships (Robertson et al. 2001). For example, in the
hormone maintained spermatogonial populations context of androgenprogestin-based male hormonal
and their progression into meiosis in gonadotropin- contraception (the efficacy of which depends upon pro-
suppressed humans (Matthiesson et al. 2006), and found gonadotropin suppression), men with sperm
stimulated spermatogonial numbers in gonadotro- concentrations below 0.1 million/ml had significantly
pin-suppressed monkeys (Weinbauer et al. 1991; lower gonadotropin levels (serum FSH 0.12 IU/l;
Marshall et al. 1995). serum LH 0.05 IU/l) than oligozoospermic men
Observations on various transgenic mouse models (sperm concentrations, 0.15 million/ml; serum FSH,
reveal that FSH supports other aspects of spermato- 0.230.5 IU/l; serum LH, 0.050.56 IU/l). In multivari-
genesis that, when combined with testosterone action, ate analysis, the suppression of serum LH to less than
are essential for quantitatively normal spermatogen- 5% of baseline values (<0.15 IU/l) was found to be a
esis (also see Section 6.6.2). Such aspects include a consistent and highly significant predictor of suppres-
possible FSH-mediated paracrine factor(s) from Ser- sion to below 1 million/ml; while that was not true for
toli cells that influences Leydig cell number, matur- serum FSH (McLachlan et al. 2004).
ation and steroidogenesis (OShaughnessy et al. 2009). Finally, it is interesting to note that seasonally
Although meiosis and spermiogenesis are regarded as breeding animals seem to have a greater dependence
androgen-dependent processes, FSH likely has some on FSH than non-seasonal animals. In the Djungarian
permissive role in these processes via its effects on hamster, the testes undergo marked involution when
Sertoli cell function (e.g. Abel et al. 2008; reviewed in the light : dark exposure is shifted from 16 : 8 to 8 : 16;
Ruwanpura et al. 2010). LH administration is unable to restore spermato-
Follicle-stimulating hormone is clearly important genesis; however, FSH reinitiates the entire process
for spermatogenesis in non-human primates and (Lerchl et al. 1993). Similarly in another seasonally
humans, as reviewed elsewhere (Nieschlag et al. breeding mammal, the prairie dog (Cynomys ludovi-
1999). For example, in the GnRH antagonist-treated canus), FSH but not LH/testosterone induced germ
non-human primate model, human FSH was able to cell activation when given during the seasonal involu-
qualitatively maintain and restore spermatogenesis tion phase (Foreman 1998).
(Weinbauer et al. 1991). Immunization against FSH In summary, FSH-independent actions are essen-
in rhesus and bonnet monkeys provoked marked tial for quantitatively normal sperm output. Import-
testicular involution and even infertility (Moudgal ant FSH-specific actions include the development of a
et al. 1997; Nieschlag et al. 1999). Studies in gonado- full complement of functional Sertoli cells with an
tropin-suppressed cynomolgus monkeys (Narula optimal ability to support germ cell development,
et al. 2002; Weinbauer et al. 2001) reveal correlations and the ability to support spermatogonial populations
between spermatogenesis and FSH. The ability of and their entry into meiosis.

139
Chapter 6: The role of testosterone in spermatogenesis

6.6.2 Androgen and follicle-stimulating Hikim and Swerdloff 1999). Germ cell survival during
gonadotropin withdrawal could be maintained by either
hormone; cooperative effects on hormone alone, but the combination of both androgen
spermatogenesis and FSH had a synergistic effect (El Shennawy et al.
It is apparent from the above discussion that andro- 1998). The ability of either hormone to prevent germ
gens and FSH have distinct roles in spermatogenesis. cell apoptosis likely explains why both FSH and hCG
However, overall these hormones act cooperatively to maintained germ cell populations in gonadotropin-
promote maximal spermatogenic output (summar- suppressed humans (Matthiesson et al. 2006).
ized in Fig. 6.5). Androgen and FSH have synergistic, When considering combined androgen and FSH
additive, and redundant effects on spermatogenesis suppression, there are species-dependent differences
and Sertoli cell activity, as demonstrated in transgenic in the response of spermatogenesis (Fig. 6.6). In
mouse models (Abel et al. 2008). Androgen and FSH rodents, gonadotropin suppression induced by GnRH
appear to cooperate by acting independently to sup- immunization causes a decline in spermatogonial
port different processes within spermatogenesis; populations (Fig. 6.6), with spermatogenesis primar-
for example FSHs ability to support spermatogonial ily arrested during meiosis (Meachem et al. 1998). In
development and testosterones ability to support contrast, in primates, spermatogenesis primarily shows
spermatid development. There is also the potential a defect in spermatogonial development, with meiotic
for androgen and FSH to cooperate in some meta- and spermatid populations largely maintained until
bolic pathways (Walker and Cheng 2005), in which they undergo a gradual attrition (Zhengwei et al.
setting either hormone may support a process to a 1998; ODonnell et al. 2001a; McLachlan et al. 2002a;
qualitative degree, but both hormones combined are Matthiesson et al. 2005). There are slight differences
required for a maximal response. The cooperative between monkeys and man in terms of the spermato-
effects of androgens and FSH on rodent and human gonial subtype first affected by gonadotropin suppres-
spermatogenesis have been reviewed recently else- sion; however, in both species there is evidence for an
where (Ruwanpura et al. 2010), and will be considered inhibition of the mitosis of Ap spermatogonia, and for
only briefly here. the transition of Ap to resting Ad spermatogonia
There is good evidence that androgen or FSH during gonadotropin suppression (McLachlan et al.
act on spermatogenesis at a lower dose when the other 2002b; Fig. 6.6). These gonadotropin suppression
is present (Zirkin 1998; McLachlan et al. 2002b; paradigms do not allow a dissection of the sensitivity
ODonnell et al. 2006). In particular there appears to of primate spermatogonia to androgen or FSH,
be a strong case for a potentiating effect of FSH making it difficult to ascribe hormone-specific roles.
on androgen action, as in many experimental settings The fact that the response of type B spermatogonia
the androgen requirements of spermatogenesis are to gonadotropin suppression correlates more closely
lower in the presence of circulating FSH (reviewed with FSH than iTT in both monkeys and men
in ODonnell et al. 2006). Conversely, studies suggest (ODonnell et al. 2001a; McLachlan et al. 2002a),
that even very low levels of androgen may facilitate together with the FSH-specific effects in rodents (see
FSH action on spermatogenesis (e.g. Chandolia et al. Section 6.6.1), suggests that these cells are particularly
1991b; Spiteri-Grech et al. 1993). Therefore the ability FSH sensitive (Fig. 6.6).
of either hormone to stimulate or maintain spermato- Postnatal Sertoli cells are a major target for andro-
genesis is greatly enhanced when even low levels of gen and FSH action, and both hormones have major
the other are present. effects on Sertoli cell morphology, function and gene
A major site of androgen and FSH cooperativity is expression (OShaughnessy et al. 2009). Sertoli cell
likely to be the prevention of apoptosis/promotion morphology and ability to support germ cells was
of germ cell survival, particularly of spermatocytes impaired in mice in which the Sertoli cells lacked
and round spermatids (reviewed in Ruwanpura et al. either active FSH or androgen receptors, but was
2010). During gonadotropin suppression, germ cell more markedly impaired in mice lacking both (Abel
apoptosis is first seen in the mid-spermatogenic stages et al. 2008). These results suggest that androgen and
(VII and VIII in rodents), with spermatocytes and FSH act in a cooperative manner to directly modulate
round spermatids being the most susceptible (Sinha Sertoli cell function, morphology and the ability to
support germ cells. The ability of androgen and FSH

140
Sertoli cells

FSH
FSH and T Essential for a full
Potentiate one anothers action complement of Sertoli cells
on Sertoli cell function and germ
cell development
Likely have complementary, T
redundant, additive and Action on Sertoli cell AR
synergistic functions within essential for normal function
Sertoli cells
Action on PTM AR supports
Sertoli cell number

Germ cells

FSH
Stimulates and maintains
spermatogonial populations
Sgonia
FSH and T
Synergistic/additive effects
on entry into meiosis

Preleptotene
scyte
FSH and T
Synergistic effects on germ cell
survival (esp. spermatocytes
and round spermatids)

Pachytene
scyte

T
Essential for completion
Early
of meiosis
round
ST

T
Essential for transition
between round and
elongating spermatids
FSH and T
Synergistic effect on
spermiation
Early
elongating
ST

Elongated
ST
Fig. 6.5 Roles of androgen and FSH in spermatogenesis as assessed in rodent models. The action of both androgen and FSH is
required for quantitatively normal spermatogenesis. This diagram briefly summarizes the likely independent and additive/synergistic effects
of androgen and FSH on spermatogenesis in rodents. Androgen and FSH act on receptors in Sertoli cells to support spermatogenesis, but
androgen also acts on other testicular somatic cells including peritubular myoid cells (PTM) and Leydig cells. The precise sites of action of
androgen and FSH in spermatogenesis have been primarily elucidated in transgenic mouse models and adult rat suppression and
reinitiation/maintenance models. Abbreviations: T, testosterone; sgonia, spermatogonia; scyte, spermatocyte; ST, spermatid.
Chapter 6: The role of testosterone in spermatogenesis

Fig. 6.6 Response of spermatogenesis


T/FSH to androgen and FSH withdrawal in
rodents and primates. Graphs show germ
cell populations in humans, monkeys and
Ad rats undergoing 1214 weeks of
100 T/FSH gonadotropin suppression. Gonadotropin
Human suppression was induced by 12 weeks of
Mitosis
Germ cells (% control)

80 testosterone plus progestin treatment in


Ap humans (see McLachlan et al. 2002a), 14
60 weeks of testosterone treatment in
Spermatogonia monkeys (see ODonnell et al. 2001a) and
40 12 weeks of immunization against GnRH
FSH? in rats (see Meachem et al. 1998). See the
cited papers for details of enumeration of
20
Mitosis germ cells. Humans and monkeys show
B similar patterns of suppression, with a
0 major block between A and
-Z

B spermatogonia, yet maintenance of


A
B

PS

T
ST

Sp T
m
rS

eS
PI

er
eI

some meiotic and spermiogenic


100 populations; whereas rat
Monkey
LH and FSH suppression (1214 wks)

spermatogenesis shows a major block


Germ cells (% control)

80 during meiosis. The cells shown to the


right are human germ cells, indicating
60 the process of spermatogenesis in man.
The specific spermatogenic processes
PS that are impaired by 1214 weeks of
40
Meiosis gonadotropin suppression in humans
(and monkeys) are indicated (T/FSH).
20 In this setting of reduced FSH and iTT,
some maintenance of meiosis and
0 spermiogenesis occurs (see graphs);
however, based on studies in rodents
-Z
A
B

PS

T
ST

Sp T
m
rS

eS
PI

er
eI

(see Section 6.5.2), it is possible that more


100 profound suppression of intratesticular
Rat androgens could impair these processes.
Germ cells (% control)

Meiotic The mitoses of Ap spermatogonia are


80 division thought to be impaired by gonadotropin
withdrawal, with the transition of Ap
60 spermatogonia to resting Ad
spermatogonia seen during acute
40 gonadotropin suppression (reviewed in
McLachlan et al. 2002b). Correlations
20 between type B spermatogonia numbers
and FSH, but not iTT, suggest that the
rST mitosis of these cells may be more sensitive
0
Spermiogenesis to FSH. The release of mature elongated
PI

Z
PS

ST

T
A

T
L-

eS

spermatids from the Sertoli cells during


rS
B-

eI

spermiation is also highly sensitive to


gonadotropin suppression in men (as well
as in monkeys and rats). Abbreviations: Ad,
type A dark spermatogonia; Ap, type A pale
rST spermatogonia; B, type B spermatogonia;
Spermiation Pl, preleptotene spermatocytes; L, leptotene
spermatocytes; Z, zygotene spermatocytes;
PS, pachytene spermatocytes; rST, round
T/FSH elST spermatids; elST, elongating spermatids;
eST eST, elongated spermatids.

to cooperate in modulating Sertoli cell function had a synergistic effect, such that approximately 50%
is also highlighted by studies investigating the process of the spermatids failed to be released (Saito et al.
of spermiation. When either androgen or FSH was 2000). Taken together, it seems likely that androgen
suppressed in adult rats, Sertoli cells failed to release and FSH regulate distinct and overlapping functions
approximately 10% of elongated spermatids. How- in Sertoli cells which cooperate to support different
ever, the suppression of both hormones in combination aspects of germ cell development.

142
Chapter 6: The role of testosterone in spermatogenesis

In summary, it can be concluded that, in rodents directly on testicular germ cells via a non-genomic
and primates, the combination of androgen and FSH pathway mediated by a G-protein-coupled receptor
consistently produces a better stimulatory effect on known as GPR30 (Chimento et al. 2010; Carreau
spermatogenesis than either factor alone. For quanti- et al. 2011). Second, efferent ductule development
tatively normal spermatogenesis and fertility, the and function was shown to be regulated by estra-
action of both androgen and FSH is required. diol-independent activation of ERa (Sinkevicius
et al. 2009), explaining why efferent ductule function
6.6.3 Aromatization of testosterone was not compromised in aromatase-deficient mice.
Third, mice expressing a mutated ERa which is
to estradiol; role in spermatogenesis unable to bind estradiol (ENERKI mice) had a sper-
The metabolism of testosterone to estradiol by the matogenic phenotype almost identical to the ArKO
aromatase enzyme and its role in spermatogenesis mouse, including a late-onset phenotype (Sinkevicius
has been the subject of intensive review, and we refer et al. 2009), suggesting that estradiol signaling via
the reader to previous reviews on this topic (ODonnell ERa is important for the progression of spermatogen-
et al. 2001b; Hess 2003; Carreau and Hess 2010; esis. This latter proposition was proven by the fact
Carreau et al. 2011). There is a variety of evidence that the administration of a selective agonist capable
to suggest that exogenous estrogens and phytoestro- of activating the mutant ERa in ENERKI mice res-
gens can exert effects during testicular development cued the spermatogenic phenotype (Sinkevicius et al.
which can have adverse consequences for male fertility 2009). Intriguingly, activation of ERa during the neo-
(reviewed in ODonnell et al. 2001b); however this natal period, but not after puberty, was required to
section will focus on the potential role of the metab- rescue the phenotype, suggesting that estradiol-
olism of testosterone to estradiol in normal dependent activation of ERa in the neonatal testis is
spermatogenesis. required for fertility in adulthood. It is not yet known
Mice lacking the aromatase gene (Robertson et al. what cells/processes in the neonatal testis rely on
1999; Robertson et al. 2002; Carreau and Hess 2010) estradiolERa action, and how they can influence
and ERa are infertile (Eddy et al. 1996; Hess et al. the maintenance of spermatogenesis in adulthood.
1997), highlighting a critical role for estrogen in Taken together, these new findings suggest that
male fertility. The disturbed spermatogenesis spermatogenesis relies on estradiol-dependent and
observed in the ERa knock-out (ERaKO) mouse independent activation of the classic estrogen recep-
was a consequence of disturbed efferent ductule tors during development, and that estradiol can exert
function, whereby the failure of the efferent ductules effects on spermatogenesis via non-classic estrogen
to resorb seminiferous tubule fluid resulted in fluid receptors (reviewed in Carreau and Hess 2010;
accumulation in the testis and secondary defects to Carreau et al. 2011).
spermatogenesis (Eddy et al. 1996; Hess et al. 1997). The ability of estradiol to stimulate spermato-
Estrogen receptor-a regulates epithelial cell morph- genesis during gonadotropin deficiency has been
ology as well as ion channel and aquaporin water the subject of some debate. Treatment of postnatal
channel function in the efferent ductules (reviewed hypogonadal (hpg) mice with estradiol stimulated
in Carreau and Hess 2010). Conversely, there was no qualitatively normal spermatogenesis, but was also
evidence of disturbed efferent ductule function in the associated with a stimulation of FSH (Ebling et al.
aromatase knock-out mouse (ArKO), but the sperm- 2000). Spermatogenesis (and FSH) was able to be
atogenesis phenotype observed was consistent with stimulated by an ERa, but not ERb, agonist (Allan
an effect on Sertoli cells and/or germ cells, and et al. 2010). Since FSH in the absence of androgen can
worsened with age (Robertson et al. 1999; 2002). only stimulate pre-meiotic germ cell development in
How this action was mediated was unclear since these mice (Haywood et al. 2003), the induction
ERbKO mice were fertile (Dupont et al. 2000) des- of spermatogenesis observed during estradiol treat-
pite ERb being widely expressed in the seminiferous ment is unlikely to be solely due to circulating FSH,
epithelium. although an FSH effect is possible (Baines et al. 2008).
Several recent advances have shed light on how The ability of estradiol to stimulate spermatogenesis
estradiol and its receptors influence spermatogenesis. relies on the presence of a functional AR, despite no
First, it has been established that estradiol can act detectable increase in iTT (Baines et al. 2005; Lim

143
Chapter 6: The role of testosterone in spermatogenesis

et al. 2008). The stimulatory effects of estradiol does not appear achievable with strategies based on
required a functional ERa, but not ERb, and were pituitary gonadotropin inhibition using GnRH
associated with changes in Sertoli cell morphology analogs and/or sex steroid administration, presum-
and gene expression, including the expression of Ser- ably because of LH-independent androgen secretion
toli cell androgen-dependent genes (Allan et al. 2010). maintaining low iTT levels (12% baseline) and some
Thus, estradiol stimulates spermatogenesis via mech- spermatogenesis in a minority ( 5%) of men. This
anisms that require both ERa and AR in a setting so-called non-responder group may well represent
of circulating FSH. It is possible that estrogen acts the extreme end of a normal distribution for andro-
directly on ERa and/or on hetereodimers of ERa/AR gen action threshold on spermatogenesis wherein a
in Sertoli cells (Allan et al. 2010) to modulate key little bit of androgen goes a long way. As argued
Sertoli cell functions which, in concert with FSH, above, the true elimination of androgen action, per-
facilitate germ cell development. While iTT levels haps through testis-specific enzyme inhibition or
are very low in this model (Baines et al. 2005; Lim targeted blockade of testicular AR or its co-modula-
et al. 2008), DHT levels were not measured, and thus tors, may well achieve the long-held goal of universal
it is possible that low levels of androgen action could azoospermia. However, this demands a better under-
be preserved via an FSH-mediated upregulation of standing of these underlying processes and the gener-
5a-reductase (Pratis et al. 2003). ation of novel therapeutic tools; this will be a long
In summary, estradiol likely has a permissive/ road and demands the continued partnership between
supportive role in spermatogenesis, although the basic and clinical scientists, and industry.
precise estradiol-dependent processes are unknown.
6.8 Key messages
6.7 Implications for treatment of  Testosterone is essential for spermatogenesis and
able to act alone to qualitatively initiate, maintain
secondary hypogonadism and for male and reinitiate spermatogenesis and formation
of spermatozoa.
hormonal contraception  Full, quantitatively normal spermatogenesis
The pivotal role of androgen action in supporting requires the combined action of testosterone and
spermatogenesis underscores certain aspects of FSH.
approaches to fertility control. First, hCG as an LH  Testosterone and FSH cooperate in the
substitute has proven effective when given at doses of regulation of spermatogenesis via
1500 IU two to three times per week. These doses redundant, additive, complementary and
appear supraphysiological when one looks at reports synergistic actions.
of iTT level with graded hCG exposure, wherein 500  Androgen actions on receptors in Sertoli cells,
peritubular myoid cells and Leydig cells, but not
IU twice weekly would seem more than adequate
germ cells, are essential for spermatogenesis.
(Roth et al. 2010b). Yet conventional (likely excess)
 Key androgen-dependent processes in rodent
hCG doses do not appear to adversely affect the spermatogenesis include the completion of
therapeutic effect, despite the provision of excess sub- meiosis and the progression through
strate for aromatization to estradiol. The hCG does spermiogenesis.
need to be titrated to avoid excessive androgen levels  A very narrow dose-response range exists
with clinical side-effects, and co-suppression of between iTT levels and sperm output; this hair
endogenous FSH (if present). Those issues aside, trigger effect combined with interindividual
there appears to be no downside to supraphysiolo- differences leads to difficulties in correlating the
gical iTT levels induced by conventional hCG doses. two parameters.
Finally, it must be emphasized that, in some men,  Testosterone metabolism to DHT may play a role
in maintaining androgen action when
hCG treatment must be protracted (one to two years),
testosterone concentrations are low.
especially for the initial induction of spermatogenesis
 Testosterone metabolism to estradiol has a
in Kallmann syndrome, but this seems only reason- supportive role in spermatogenesis, though
able given the time course of normal puberty. the precise cellular processes involved are
Conversely in the male hormonal contraception unclear.
paradigm, the complete abolition of androgen effect

144
Chapter 6: The role of testosterone in spermatogenesis

 Several androgen-dependent genes have been effective, and co-administration with FSH takes
identified in the testis, but the cellular pathways advantage of the well-known synergies at
by which androgens mediate somatic and germ multiple points in germ cell development
cell functions are unclear. and spermiation.
 For male hormonal contraception, withdrawal of
androgen action is essential to achieving
maximal effect. In some men, LH-independent
Leydig cell androgen secretion apparently
Acknowledgements
maintains spermatogenesis, pointing to the need The authors were supported by a NHMRC Australia
for alternative approaches. Program Grant no. 494802 and by the Victorian Gov-
 In the setting of hypogonadotropic hypogonadism, ernments Operational Infrastructure Support Program.
hCG (as an LH substitute) restores iTT, but R. I. McLachlan was supported by a NHMRC Australia
treatment may take monthsyears to be Research Fellowship no. 441103.

6.9 References testosterone. J Clin Endocrinol


Metab 81:902908
Health and Dysfunction, 3rd edn.
Springer-Verlag, Berlin, pp 169192
Abel MH, Baker PJ, Charlton HM,
Monteiro A, Verhoeven G, De Awoniyi CA, Santulli R, Sprando RL, Bernard DJ, Fortin J, Wang Y, Lamba P
Gendt K, Guillou F, OShaughnessy Ewing LL, Zirkin BR (1989) (2010) Mechanisms of FSH
PJ (2008) Spermatogenesis and Restoration of advanced synthesis: what we know, what we
Sertoli cell activity in mice spermatogenic cells in the dont, and why you should care.
lacking Sertoli cell receptors for experimentally regressed rat testis: Fertil Steril 93:24652485
follicle-stimulating hormone quantitative relationship to
Bckers TM, Nieschlag E, Kreutz MR,
and androgen. Endocrinology testosterone concentration
Bergmann M (1994) Localization of
149:32793285 within the testis. Endocrinology
follicle-stimulating hormone (FSH)
124:12171223
Allan CM, Garcia A, Spaliviero J, immunoreactivity and hormone
Awoniyi CA, Zirkin BR, receptor mRNA in testicular tissue
Zhang FP, Jimenez M, Huhtaniemi Chandrashekar V, Schlaff WD
I, Handelsman DJ (2004) of infertile men. Cell Tissue Res
(1992) Exogenously administered 278:595600
Complete Sertoli cell proliferation testosterone maintains
induced by follicle-stimulating spermatogenesis quantitatively in Bremner WJ, Millar MR, Sharpe RM,
hormone (FSH) independently adult rats actively immunized Saunders PT (1994)
of luteinizing hormone against gonadotropin-releasing Immunohistochemical localization
activity: evidence from genetic hormone. Endocrinology of androgen receptors in the rat
models of isolated FSH action. 130:32833288 testis: evidence for stage-dependent
Endocrinology 145:15871593 expression and regulation by
Baines H, Nwagwu MO, Furneaux EC, androgens. Endocrinology
Allan CM, Couse JF, Simanainen U, Stewart J, Kerr JB, Mayhew TM, 135:12271234
Spaliviero J, Jimenez M, Rodriguez Ebling FJ (2005) Estrogenic
K, Korach KS, Handelsman DJ induction of spermatogenesis in the Bchter D, Behre HM, Kliesch S,
(2010) Estradiol induction of hypogonadal (hpg) mouse: role of Nieschlag E (1998) Pulsatile GnRH
spermatogenesis is mediated via an androgens. Reproduction or human chorionic gonadotropin/
estrogen receptor-alpha mechanism 130:643654 human menopausal gonadotropin
involving neuroendocrine as effective treatment for men with
Baines H, Nwagwu MO, Hastie GR,
activation of follicle-stimulating hypogonadotropic hypogonadism: a
Wiles RA, Mayhew M, Ebling FJ
hormone secretion. Endocrinology review of 42 cases. Eur J Endocrinol
(2008) Effects of estradiol and FSH
151:28002810 139:298303
on maturation of the testis in the
Anderson RA, Wallace AM, Wu FC hypogonadal (hpg) mouse. Reprod Burris AS, Rodbard HW, Winters SJ,
(1996) Comparison between Biol Endocrinol 6:4 Sherins RJ (1988) Gonadotropin
testosterone enanthate-induced therapy in men with isolated
Behre HM, Nieschlag E, Weiacker P,
azoospermia and oligozoospermia hypogonadotropic hypogonadism:
Simoni M, Partsch CJ (2010)
in a male contraceptive study. III. the response to human chorionic
Diseases of the hypothalamus and
Higher 5 alpha-reductase activity in gonadotropin is predicted by initial
the pituitary gland. In: Nieschlag E,
oligozoospermic men administered testicular size. J Clin Endocrinol
Behre HM, Nieschlag S (eds)
supraphysiological doses of Metab 66:11441151
Andrology. Male Reproductive

145
Chapter 6: The role of testosterone in spermatogenesis

Cameron DF, Muffly KE (1991) Clermont Y, Leblond CP (1955) Morrison JR, Loveland KL, Hedger
Hormonal regulation of Spermiogenesis of man, monkey, MP (2004) The role of activin,
spermatid binding. J Cell Sci ram and other mammals as shown follistatin and inhibin in testicular
100:623633 by the periodic acid-Schiff physiology. Mol Cell Endocrinol
technique. Am J Anat 96:229253 225:5764
Carreau S, Hess RA (2010) Oestrogens
and spermatogenesis. Philos Corpechot C, Baulieu EE, Robel P Depenbusch M, von Eckardstein S,
Trans R Soc Lond B Biol Sci (1981) Testosterone, Simoni M, Nieschlag E (2002)
365:15171535 dihydrotestosterone and Maintenance of spermatogenesis in
androstanediols in plasma, testes hypogonadotropic hypogonadal
Carreau S, Bois C, Zanatta L, Silva FR,
and prostates of rats during men with human chorionic
Bouraima-Lelong H, Delalande C
development. Acta Endocrinol gonadotropin alone. Eur
(2011) Estrogen signaling in
(Copenh) 96:127135 J Endocrinol 147:617624
testicular cells. Life Sci 89:584587
Dankbar B, Brinkworth MH, Schlatt S, de Rooij DG, Grootegoed JA (1998)
Chandolia RK, Weinbauer GF, Behre
Weinbauer GF, Nieschlag E, Spermatogonial stem cells. Curr
HM, Nieschlag E (1991a) Opin Cell Biol 10:694701
Gromoll J (1995) Ubiquitous
Evaluation of a peripherally
expression of the androgen Deslypere JP, Young M, Wilson JD,
selective antiandrogen (Casodex) as
receptor and testis-specific McPhaul MJ (1992) Testosterone
a tool for studying the relationship
expression of the FSH receptor in and 5 alpha-dihydrotestosterone
between testosterone and
the cynomolgus monkey (Macaca interact differently with the
spermatogenesis in the rat. J Steroid
fascicularis) revealed by a androgen receptor to enhance
Biochem Mol Biol 38:367375
ribonuclease protection assay. transcription of the MMTV-CAT
Chandolia RK, Weinbauer GF, J Steroid Biochem Mol Biol reporter gene. Mol Cell Endocrinol
Fingscheidt U, Bartlett JM, 55:3541 88:1522
Nieschlag E (1991b) Effects of
De Franca LR, Hess RA, Cooke PS, Dupont S, Krust A, Gansmuller A,
flutamide on testicular involution
Russell LD (1995) Neonatal Dierich A, Chambon P, Mark M
induced by an antagonist of
hypothyroidism causes delayed (2000) Effect of single and
gonadotrophin-releasing hormone
Sertoli cell maturation in rats compound knockouts of estrogen
and on stimulation of
treated with propylthiouracil: receptors alpha (ERalpha) and beta
spermatogenesis by follicle-
evidence that the Sertoli cell (ERbeta) on mouse reproductive
stimulating hormone in rats.
controls testis growth. Anat Rec phenotypes. Development 127:4277
J Reprod Fertil 93:313323
242:5769 4291
Chang C, Chen YT, Yeh SD, Xu Q,
De Gendt K, Swinnen JV, Saunders PT, Ebling FJ, Brooks AN, Cronin AS, Ford
Wang RS, Guillou F, Lardy H, Yeh S
Schoonjans L, Dewerchin M, Devos A, H, Kerr JB (2000) Estrogenic
(2004) Infertility with defective
Tan K, Atanassova N, Claessens F, induction of spermatogenesis in the
spermatogenesis and
Lecureuil C, Heyns W, Carmeliet P, hypogonadal mouse. Endocrinology
hypotestosteronemia in male mice
lacking the androgen receptor in
Guillou F, Sharpe RM, Verhoeven G 141:28612869
(2004) A Sertoli cell-selective
Sertoli cells. Proc Natl Acad Sci USA Eddy EM, Washburn TF, Bunch DO,
knockout of the androgen receptor
101:68766881 Goulding EH, Gladen BC, Lubahn
causes spermatogenic arrest in
Chapin RE, Wine RN, Harris MW, DB, Korach KS (1996) Targeted
meiosis. Proc Natl Acad Sci USA
disruption of the estrogen receptor
Borchers CH, Haseman JK (2001) 101:13271332
Structure and control of a cell-cell gene in male mice causes alteration
de Kretser DM, Kerr JB (1988) The of spermatogenesis and infertility.
adhesion complex associated
with spermiation in rat
cytology of the testis. In: Knobil E, Endocrinology 137:47964805
Neill JD (eds) The Physiology of
seminiferous epithelium. J Androl El Shennawy A, Gates RJ, Russell LD
Reproduction. Raven Press Ltd, New
22:10301052 (1998) Hormonal regulation of
York, pp 837932
Chimento A, Sirianni R, Delalande C, spermatogenesis in the
Silandre D, Bois C, Ando S, de Kretser DM, Loveland KL, Meehan hypophysectomized rat: cell
Maggiolini M, Carreau S, Pezzi V T, OBryan MK, Phillips DJ, viability after hormonal
(2010) 17 beta-estradiol activates Wreford NG (2001) Inhibins, replacement in adults after
rapid signaling pathways involved activins and follistatin: actions on intermediate periods of
in rat pachytene spermatocytes the testis. Mol Cell Endocrinol hypophysectomy. J Androl
apoptosis through GPR30 and ER 180:8792 19:320334; discussion:341342
alpha. Mol Cell Endocrinol de Kretser DM, Buzzard JJ, Okuma Y, Fingscheidt U, Weinbauer GF, Fehm
320:136144 OConnor AE, Hayashi T, Lin SY, HL, Nieschlag E (1998) Regulation

146
Chapter 6: The role of testosterone in spermatogenesis

of gonadotrophin secretion by Gromoll J, Wistuba J, Terwort N, www.endotext.org/male/male2/


inhibin, testosterone and Godmann M, Muller T, Simoni M maleframe2.htm (MDText.com,
gonadotrophin-releasing hormone (2003) A new subclass of the Inc), South Dartmouth, MA,
in pituitary cell cultures of luteinizing hormone/chorionic Chapter 5
male monkeys. J Endocrinol gonadotropin receptor lacking exon
Haywood M, Tymchenko N, Spaliviero
159:103110 10 messenger RNA in the New J, Koch A, Jimenez M, Gromoll J,
Finkel DM, Phillips JL, Snyder PJ World monkey (Platyrrhini)
Simoni M, Nordhoff V,
(1985) Stimulation of lineage. Biol Reprod 69:7580 Handelsman DJ, Allan CM (2002)
spermatogenesis by gonadotropins Grover A, Sairam MR, Smith CE, An activated human follicle-
in men with hypogonadotropic Hermo L (2004) Structural and stimulating hormone (FSH)
hypogonadism. N Engl J Med functional modifications of Sertoli receptor stimulates FSH-like activity
313:651655 cells in the testis of adult follicle- in gonadotropin-deficient
Foreman D (1998) Effects of exogenous stimulating hormone receptor transgenic mice. Mol Endocrinol
hormones on spermatogenesis in knockout mice. Biol Reprod 16:25822591
the male prairie dog (Cynomys 71:117129 Haywood M, Spaliviero J, Jimemez M,
ludovicianus). Anat Rec Grumbach MM (2005) A window of King NJ, Handelsman DJ, Allan CM
250:4561 opportunity: the diagnosis of (2003) Sertoli and germ cell
Franca LR, Ogawa T, Avarbock MR, gonadotropin deficiency in the male development in hypogonadal (hpg)
Brinster RL, Russell LD (1998) infant. J Clin Endocrinol Metab mice expressing transgenic follicle-
Germ cell genotype controls cell 90:31223127 stimulating hormone alone or in
cycle during spermatogenesis in the Guo Q, Kumar TR, Woodruff T, combination with testosterone.
rat. Biol Reprod 59:13711377 Hadsell LA, DeMayo FJ, Matzuk Endocrinology 144:509517
Gendrel D, Chaussain JL, Roger M, Job MM (1998) Overexpression of Heckert L, Griswold MD (1993)
JC (1980) Simultaneous mouse follistatin causes Expression of the FSH receptor in
postnatal rise of plasma LH and reproductive defects in transgenic the testis. Recent Prog Horm Res
testosterone in male infants. mice. Mol Endocrinol 12:96106 48:6177
J Pediatr 97:600602 Hall JM, McDonnell DP (1999) The Hess RA (2003) Estrogen in the adult
George FW, Johnson L, Wilson JD estrogen receptor beta-isoform male reproductive tract: a review.
(1989) The effect of a 5 alpha- (ERbeta) of the human estrogen Reprod Biol Endocrinol 1:52
reductase inhibitor on androgen receptor modulates ERalpha
Hess RA, Bunick D, Lee KH, Bahr J,
physiology in the immature male transcriptional activity and is a key
Taylor JA, Korach KS, Lubahn DB
rat. Endocrinology 125:24342438 regulator of the cellular response to
(1997) A role for oestrogens in the
Grino PB, Griffin JE, Wilson JD (1990) estrogens and antiestrogens.
male reproductive system. Nature
Endocrinology 140:55665578
Testosterone at high concentrations 390:509512
interacts with the human androgen Handelsman DJ (2008) Androgen
Holdcraft RW, Braun RE (2004a)
receptor similarly to physiology, pharmacology and
Androgen receptor function is
dihydrotestosterone. Endocrinology abuse. In: de Groot L, McLachlan RI
required in Sertoli cells for the
126:11651172 (eds) Endocrinology of Male
terminal differentiation of
Gromoll J, Partsch CJ, Simoni M, Reproduction. www.endotext.org/
haploid spermatids. Development
male/male2/maleframe2.htm
Nordhoff V, Sippell WG, Nieschlag 131:459467
E, Saxena BB (1998) A mutation in (MDText.com, Inc), South
Dartmouth, MA, Chapter 2 Holdcraft RW, Braun RE (2004b)
the first transmembrane domain of
Hormonal regulation of
the lutropin receptor causes male Handelsman DJ, Spaliviero JA,
spermatogenesis. Int J Androl
precocious puberty. J Clin Simpson JM, Allan CM, Singh J
27:335342
Endocrinol Metab 83:476480 (1999) Spermatogenesis without
Gromoll J, Eiholzer U, Nieschlag E, gonadotropins: maintenance Hu Z, Dandekar D, OShaughnessy PJ,
Simoni M (2000) Male has a lower testosterone threshold De Gendt K, Verhoeven G,
hypogonadism caused by than initiation. Endocrinology Wilkinson MF (2010) Androgen-
homozygous deletion of exon 10 of 140:39383946 induced Rhox homeobox genes
the luteinizing hormone (LH) Hayes F, Pitteloud N (2004) modulate the expression of AR-
receptor: differential action of Hypogonadotropic hypogonadism regulated genes. Mol Endocrinol
human chorionic gonadotropin and and gonadotropin therapy. In: de 24:6075
LH. J Clin Endocrinol Metab Groot L, McLachlan RI (eds) Huang HF, Marshall GR, Rosenberg R,
85:22812286 Endocrinology of Male Reproduction. Nieschlag E (1987) Restoration of

147
Chapter 6: The role of testosterone in spermatogenesis

spermatogenesis by high levels of cells in vitro. Reproduction (Phodopus sungorus). Biol Reprod
testosterone in hypophysectomized 133:11691179 49:11081116
rats after long-term regression. Kangasniemi M, Dodge K, Pemberton Lim P, Allan CM, Notini AJ, Axell AM,
Acta Endocrinol (Copenh) AE, Huhtaniemi I, Meistrich ML Spaliviero J, Jimenez M, Davey R,
116:433444 (1996) Suppression of mouse McManus J, MacLean HE, Zajac JD,
Jarow JP, Zirkin BR (2005) The spermatogenesis by a gonadotropin- Handelsman DJ (2008) Oestradiol-
androgen microenvironment of the releasing hormone antagonist and induced spermatogenesis requires a
human testis and hormonal control antiandrogen: failure to protect functional androgen receptor.
of spermatogenesis. Ann N Y Acad against radiation-induced Reprod Fertil Dev 20:861870
Sci 1061:208220 gonadal damage. Endocrinology
Lim P, Robson M, Spaliviero J,
Jarow JP, Chen H, Rosner TW, 137:949955
McTavish KJ, Jimenez M, Zajac JD,
Trentacoste S, Zirkin BR (2001) Killian J, Pratis K, Clifton RJ, Stanton Handelsman DJ, Allan CM (2009)
Assessment of the androgen PG, Robertson DM, ODonnell L Sertoli cell androgen receptor DNA
environment within the human (2003) 5alpha-reductase isoenzymes binding domain is essential for the
testis: minimally invasive method to 1 and 2 in the rat testis during completion of spermatogenesis.
obtain intratesticular fluid. J Androl postnatal development. Biol Reprod Endocrinology 150:47554765
22:640645 68:17111718
Lindsey JS, Wilkinson MF (1996) Pem:
Jeyaraj DA, Grossman G, Petrusz P Kinniburgh D, Anderson RA, Baird a testosterone- and LH-regulated
(2003) Dynamics of testicular germ DT (2001) Suppression of homeobox gene expressed in mouse
cell apoptosis in normal mice and spermatogenesis with desogestrel Sertoli cells and epididymis. Dev
transgenic mice overexpressing rat and testosterone pellets is not Biol 179:471484
androgen-binding protein. Reprod enhanced by addition of finasteride.
Liu PY, Gebski VJ, Turner L, Conway
Biol Endocrinol 1:48 J Androl 22:8895
AJ, Wishart SM, Handelsman DJ
Johnsen SG (1978) Maintenance of Kolho KL, Huhtaniemi I (1989a) (2002) Predicting pregnancy and
spermatogenesis induced by HMG Neonatal treatment of male rats spermatogenesis by survival analysis
treatment by means of continuous with a gonadotropin-releasing during gonadotrophin treatment of
HCG treatment in hormone antagonist results in gonadotrophin-deficient infertile
hypogonadotrophic men. Acta altered function of the pituitary- men. Hum Reprod 17:625633
Endocrinol (Copenh) 89:763769 testicular axis in adult age. Biol
Liu PY, Swerdloff RS, Anawalt BD,
Johnson L, McKenzie KS, Snell JR Reprod 41:10841090
Anderson RA, Bremner WJ, Elliesen
(1996) Partial wave in human Kolho KL, Huhtaniemi I (1989b) J, Gu YQ, Kersemaekers WM,
seminiferous tubules appears to be a Suppression of pituitary-testis McLachlan RI, Meriggiola MC,
random occurrence. Tissue Cell function in rats treated neonatally Nieschlag E, Sitruk-Ware R,
28:127136 with a gonadotrophin-releasing Vogelsong K, Wang XH, Wu FC,
Johnston DS, Russell LD, Friel PJ, hormone agonist and antagonist: Zitzmann M, Handelsman DJ,
Griswold MD (2001) Murine germ acute and long-term effects. Wang C (2008) Determinants of the
cells do not require functional J Endocrinol 123:8391 rate and extent of spermatogenic
androgen receptors to complete Kremer H, Kraaij R, Toledo SP, Post suppression during hormonal male
spermatogenesis following M, Fridman JB, Hayashida CY, van contraception: an integrated
spermatogonial stem cell Reen M, Milgrom E, Ropers HH, analysis. J Clin Endocrinol Metab
transplantation. Endocrinology Mariman E (1995) Male 93:17741783
142:24052408 pseudohermaphroditism due to a Liu PY, Baker HW, Jayadev V,
Johnston H, Baker PJ, Abel M, homozygous missense Zacharin M, Conway AJ,
Charlton HM, Jackson G, Fleming mutation of the luteinizing Handelsman DJ (2009) Induction of
L, Kumar TR, OShaughnessy PJ hormone receptor gene. Nat Genet spermatogenesis and fertility during
(2004) Regulation of Sertoli cell 9:160164 gonadotropin treatment of
number and activity by follicle- Lerchl A, Sotiriadou S, Behre HM, gonadotropin-deficient infertile
stimulating hormone and androgen Pierce J, Weinbauer GF, Kliesch S, men: predictors of fertility outcome.
during postnatal development in the Nieschlag E (1993) Restoration of J Clin Endocrinol Metab 94:801808
mouse. Endocrinology 145:318329 spermatogenesis by follicle- Loss ES, Jacobsen M, Costa ZS, Jacobus
Kaituu-Lino TJ, Sluka P, Foo CF, stimulating hormone despite low AP, Borelli F, Wassermann GF
Stanton PG (2007) Claudin-11 intratesticular testosterone in (2004) Testosterone modulates K(+)
expression and localisation is photoinhibited hypogonadotropic ATP channels in Sertoli cell
regulated by androgens in rat Sertoli Djungarian hamsters membrane via the PLC-PIP2

148
Chapter 6: The role of testosterone in spermatogenesis

pathway. Horm Metab Res administration in normal men: in the adult rat. Biol Reprod
36:519525 evidence that follicle-stimulating 51:945955
Lunn SF, Cowen GM, Fraser HM hormone is necessary for the McLachlan RI, Wreford NG, Tsonis C,
(1997) Blockade of the neonatal maintenance of quantitatively De Kretser DM, Robertson DM
increase in testosterone by a GnRH normal spermatogenesis in (1994b) Testosterone effects on
antagonist: the free androgen index, man. J Clin Endocrinol Metab spermatogenesis in the
reproductive capacity and 62:11841192 gonadotropin-releasing hormone-
postmortem findings in the male Matthiesson KL, Stanton PG, immunized rat. Biol Reprod
marmoset monkey. J Endocrinol ODonnell L, Meachem SJ, Amory 50:271280
154:125131 JK, Berger R, Bremner WJ, McLachlan RI, McDonald J, Rushford
Marshall GR, Wickings EJ, Ldecke McLachlan RI (2005) Effects of D, Robertson DM, Garrett C, Baker
DK, Nieschlag E (1983) Stimulation testosterone and levonorgestrel HW (2000) Efficacy and
of spermatogenesis in stalk- combined with a 5alpha-reductase acceptability of testosterone
sectioned rhesus monkeys by inhibitor or gonadotropin-releasing implants, alone or in combination
testosterone alone. J Clin Endocrinol hormone antagonist on with a 5alpha-reductase inhibitor,
Metab 57:152159 spermatogenesis and intratesticular for male hormonal contraception.
steroid levels in normal men. J Clin Contraception 62:7378
Marshall GR, Wickings EJ, Nieschlag E Endocrinol Metab 90:56475655
(1984) Testosterone can initiate McLachlan RI, ODonnell L, Stanton
spermatogenesis in an immature Matthiesson KL, McLachlan RI, PG, Balourdos G, Frydenberg M, de
nonhuman primate, Macaca ODonnell L, Frydenberg M, Kretser DM, Robertson DM (2002a)
fascicularis. Endocrinology Robertson DM, Stanton PG, Effects of testosterone plus
114:22282233 Meachem SJ (2006) The relative medroxyprogesterone acetate on
roles of follicle-stimulating semen quality, reproductive
Marshall GR, Jockenhvel F, Ldecke hormone and luteinizing hormone
D, Nieschlag E (1986) Maintenance hormones, and germ cell
in maintaining spermatogonial populations in normal young men.
of complete but quantitatively maturation and spermiation in
reduced spermatogenesis in J Clin Endocrinol Metab
normal men. J Clin Endocrinol 87:546556
hypophysectomized monkeys by Metab 91:39623969
testosterone alone. Acta Endocrinol McLachlan RI, ODonnell L, Meachem
(Copenh) 113:424431 McConnell DS, Wang Q, Sluss PM,
SJ, Stanton PG, de Kretser DM,
Bolf N, Khoury RH, Schneyer AL,
Marshall GR, Zorub DS, Plant TM Pratis K, Robertson DM (2002b)
Midgley AR Jr, Reame NE, Crowley
(1995) Follicle-stimulating Identification of specific sites of
WF Jr, Padmanabhan V (1998)
hormone amplifies the population hormonal regulation in
A two-site chemiluminescent
of differentiated spermatogonia in spermatogenesis in rats, monkeys,
assay for activin-free follistatin
the hypophysectomized and man. Recent Prog Horm Res
reveals that most follistatin
testosterone-replaced adult rhesus 57:149179
circulating in men and normal
monkey (Macaca mulatta). cycling women is in an activin- McLachlan RI, Robertson DM,
Endocrinology bound state. J Clin Endocrinol Pruysers E, Ugoni A, Matsumoto
136:35043511 Metab 83:851858 AM, Anawalt BD, Bremner WJ,
Matsumoto AM, Bremner WJ (1985) McKinnell C, Saunders PT, Fraser HM, Meriggiola C (2004) Relationship
Stimulation of sperm production by between serum gonadotropins and
Kelnar CJ, Kivlin C, Morris KD,
human chorionic gonadotropin Sharpe RM (2001) Comparison of spermatogenic suppression in men
after prolonged gonadotropin undergoing steroidal contraceptive
androgen receptor and oestrogen
suppression in normal men. receptor beta immunoexpression in treatment. J Clin Endocrinol Metab
J Androl 6:137143 89:142149
the testes of the common marmoset
Matsumoto AM, Karpas AE, Paulsen (Callithrix jacchus) from birth to Meachem SJ, Wreford NG, Stanton
CA, Bremner WJ (1983) adulthood: low androgen receptor PG, Robertson DM, McLachlan RI
Reinitiation of sperm production in immunoexpression in Sertoli cells (1998) Follicle-stimulating
gonadotropin-suppressed normal during the neonatal increase in hormone is required for the initial
men by administration of follicle- testosterone concentrations. phase of spermatogenic restoration
stimulating hormone. J Clin Invest Reproduction 122:419429 in adult rats following gonadotropin
72:10051015 McLachlan RI, Wreford NG, Meachem suppression. J Androl 19:725735
Matsumoto AM, Karpas AE, Bremner SJ, De Kretser DM, Robertson DM Means AR, Dedman JR, Tash JS,
WJ (1986) Chronic human (1994a) Effects of testosterone Tindall DJ, van Sickle M, Welsh MJ
chorionic gonadotropin on spermatogenic cell populations (1980) Regulation of the testis

149
Chapter 6: The role of testosterone in spermatogenesis

Sertoli cell by follicle stimulating to adult monkeys is related to ODonnell L, Robertson KM, Jones
hormone. Annu Rev Physiol follicle stimulating hormone ME, Simpson ER (2001b) Estrogen
42:5970 suppression and not to and spermatogenesis. Endocr Rev
Meng J, Holdcraft RW, Shima JE, intratesticular androgens. J Clin 22:289318
Griswold MD, Braun RE (2005) Endocrinol Metab 87:33993406 ODonnell L, Meachem SJ, Stanton PG,
Androgens regulate the Nicholls PK, Harrison CA, Walton KL, McLachlan RI (2006) Endocrine
permeability of the blood-testis McLachlan RI, ODonnell L, regulation of spermatogenesis. In:
barrier. Proc Natl Acad Sci USA Stanton PG (2011) Hormonal Neill JD (ed) Knobil and Neills
102:1669616700 regulation of Sertoli cell micro- Physiology of Reproduction, 3rd edn.
Meng J, Greenlee AR, Taub CJ, Braun RNAs at spermiation. Endocrinology Elsevier, San Diego, CA,
RE (2011) Sertoli cell-specific 152:16701683 pp 10171069
deletion of the androgen receptor Nieschlag E, Simoni M, Gromoll J, ODonnell L, Pratis K, Wagenfeld A,
compromises testicular immune Weinbauer GF (1999) Role of FSH Gottwald U, Muller J, Leder G,
privilege in mice. Biol Reprod in the regulation of McLachlan RI, Stanton PG (2009)
85:254260 spermatogenesis: clinical aspects. Transcriptional profiling of the
Meriggiola MC, Bremner WJ, Clin Endocrinol (Oxf) 51:139146 hormone-responsive stages of
Costantino A, Di Cintio G, spermatogenesis reveals cell-, stage-,
ODonnell L, McLachlan RI, Wreford
Flamigni C (1998) Low dose of and hormone-specific events.
NG, Robertson DM (1994)
cyproterone acetate and Endocrinology 150:50745084
Testosterone promotes the
testosterone enanthate for conversion of round spermatids ODonnell L, Nicholls PK, OBryan
contraception in men. Hum Reprod between stages VII and VIII of the MK, McLachlan RI, Stanton PG
13:12251229 rat spermatogenic cycle. (2011) Spermiation: the process of
Meriggiola MC, Costantino A, Endocrinology 135:26082614 sperm release. Spermatogenesis
Bremner WJ, Morselli-Labate AM ODonnell L, Stanton PG, Wreford 1:1435
(2002) Higher testosterone dose NG, Robertson DM, McLachlan RI OShaughnessy PJ, Morris ID,
impairs sperm suppression induced (1996a) Inhibition of 5 alpha- Huhtaniemi I, Baker PJ, Abel MH
by a combined androgen-progestin reductase activity impairs the (2009) Role of androgen and
regimen. J Androl 23:684690 testosterone-dependent restoration gonadotrophins in the development
Michel C, Gromoll J, Chandolia R, of spermiogenesis in adult rats. and function of the Sertoli cells and
Luetjens CM, Wistuba J, Simoni M Endocrinology 137:27032710 Leydig cells: data from mutant and
(2007) LHR splicing variants and ODonnell L, McLachlan RI, Wreford genetically modified mice. Mol Cell
gene expression in the marmoset NG, de Kretser DM, Robertson DM Endocrinol 306:28
monkey. Mol Cell Endocrinol (1996b) Testosterone withdrawal OShaughnessy PJ, Verhoeven G, De
279:915 promotes stage-specific detachment Gendt K, Monteiro A, Abel MH
Morse HC, Horike N, Rowley MJ, of round spermatids from the rat (2010a) Direct action through the
Heller CG (1973) Testosterone seminiferous epithelium. Biol Sertoli cells is essential for androgen
concentrations in testes of normal Reprod 55:895901 stimulation of spermatogenesis.
men: effects of testosterone ODonnell L, Pratis K, Stanton PG, Endocrinology 151:23432348
propionate administration. J Clin Robertson DM, McLachlan RI OShaughnessy PJ, Monteiro A,
Endocrinol Metab 37:882886 (1999) Testosterone-dependent Verhoeven G, De Gendt K, Abel
Moudgal NR, Sairam MR, restoration of spermatogenesis in MH (2010b) Effect of FSH on
Krishnamurthy HN, Sridhar S, adult rats is impaired by a 5alpha- testicular morphology and
Krishnamurthy H, Khan H (1997) reductase inhibitor. J Androl spermatogenesis in gonadotrophin-
Immunization of male bonnet 20:109117 deficient hypogonadal mice lacking
monkeys (M. radiata) with a ODonnell L, Narula A, Balourdos G, androgen receptors. Reproduction
recombinant FSH receptor Gu YQ, Wreford NG, Robertson 139:177184
preparation affects testicular DM, Bremner WJ, McLachlan RI Pakarainen T, Zhang FP, Makela S,
function and fertility. Endocrinology (2001a) Impairment of Poutanen M, Huhtaniemi I (2005)
138:30653068 spermatogonial development and Testosterone replacement therapy
Narula A, Gu YQ, ODonnell L, spermiation after testosterone- induces spermatogenesis and
Stanton PG, Robertson DM, induced gonadotropin suppression partially restores fertility in
McLachlan RI, Bremner WJ (2002) in adult monkeys (Macaca luteinizing hormone receptor
Variability in sperm suppression fascicularis). J Clin Endocrinol knockout mice. Endocrinology
during testosterone administration Metab 86:18141822 146:596606

150
Chapter 6: The role of testosterone in spermatogenesis

Pitteloud N, Dwyer AA, DeCruz S, Lee Simpson ER (1999) Impairment of Saito K, ODonnell L, McLachlan RI,
H, Boepple PA, Crowley WF Jr, spermatogenesis in mice lacking a Robertson DM (2000) Spermiation
Hayes FJ (2008) The relative role of functional aromatase (cyp 19) gene. failure is a major contributor to
gonadal sex steroids and Proc Natl Acad Sci USA 96: early spermatogenic suppression
gonadotropin-releasing hormone 79867991 caused by hormone withdrawal in
pulse frequency in the regulation of Robertson KM, ODonnell L, Simpson adult rats. Endocrinology
follicle-stimulating hormone ER, Jones ME (2002) The phenotype 141:27792785
secretion in men. J Clin Endocrinol of the aromatase knockout mouse Schauwaers K, De Gendt K, Saunders
Metab 93:26862692 reveals dietary phytoestrogens PT, Atanassova N, Haelens A,
Pratis K, ODonnell L, Ooi GT, Stanton impact significantly on testis Callewaert L, Moehren U, Swinnen
PG, McLachlan RI, Robertson DM function. Endocrinology 143: JV, Verhoeven G, Verrijdt G,
(2003) Differential regulation of rat 29132921 Claessens F (2007) Loss of androgen
testicular 5alpha-reductase type 1 Roth MY, Lin K, Amory JK, receptor binding to selective
and 2 isoforms by testosterone Matsumoto AM, Anawalt BD, androgen response elements
and FSH. J Endocrinol Snyder CN, Kalhorn TF, Bremner causes a reproductive phenotype
176:393403 WJ, Page ST (2010a) Serum LH in a knockin mouse model.
Ramaswamy S, Pohl CR, McNeilly AS, correlates highly with intratesticular Proc Natl Acad Sci USA
Winters SJ, Plant TM (1998) The steroid levels in normal men. 104:49614966
time course of follicle-stimulating J Androl 31:138145 Schlatt S, Weinbauer GF, Arslan M,
hormone suppression by Roth MY, Page ST, Lin K, Anawalt BD, Nieschlag E (1993) Appearance of
recombinant human inhibin A in Matsumoto AM, Snyder CN, Marck alpha-smooth muscle actin in
the adult male rhesus monkey BT, Bremner WJ, Amory JK peritubular cells of monkey testes is
(Macaca mulatta). Endocrinology (2010b) Dose-dependent increase in induced by androgens, modulated
139:34093415 intratesticular testosterone by very by follicle-stimulating hormone,
Rannikko A, Penttila TL, Zhang FP, low-dose human chorionic and maintained after hormonal
Toppari J, Parvinen M, Huhtaniemi gonadotropin in normal men with withdrawal. J Androl 14:340350
I (1996) Stage-specific expression of experimental gonadotropin Schulze W, Rehder U (1984)
the FSH receptor gene in the deficiency. J Clin Endocrinol Metab Organization and morphogenesis of
prepubertal and adult rat 95:38063813 the human seminiferous epithelium.
seminiferous epithelium. Roth MY, Page ST, Lin K, Anawalt BD, Cell Tissue Res 237:395407
J Endocrinol 151:2935 Matsumoto AM, Marck B, Bremner Sharpe RM, McKinnell C, Kivlin C,
Re M, Micali F, Santoro L, Cuomo M, WJ, Amory JK (2011) The effect of Fisher JS (2003) Proliferation and
Racheli T, Scapellato F, Iannitelli M gonadotropin withdrawal and functional maturation of Sertoli
(1979) Histological characteristics stimulation with human chorionic cells, and their relevance to
of the human testis after long-term gonadotropin on intratesticular disorders of testis function
treatment with cyproterone acetate. androstenedione and DHEA in in adulthood. Reproduction
Arch Androl 3:263268 normal men. J Clin Endocrinol 125:769784
Ren HP, Russell LD (1991) Clonal Metab 96:11751181 Shenker A, Laue L, Kosugi S,
development of interconnected Russell LD, Clermont Y (1977) Merendino JJ Jr, Minegishi T,
germ cells in the rat and its Degeneration of germ cells in Cutler GB Jr (1993) A constitutively
relationship to the segmental and normal, hypophysectomized and activating mutation of the
subsegmental organization of hormone treated luteinizing hormone receptor in
spermatogenesis. Am J Anat hypophysectomized rats. familial male precocious puberty.
192:121128 Anat Rec 187:347366 Nature 365:652654
Robertson DM, Pruysers E, Stephenson Russell LD, Ettlin RA, Sinha Hikim AP, Shetty G, Wilson G, Huhtaniemi I,
T, Pettersson K, Morton S, Clegg ED (1990) Histological and Boettger-Tong H, Meistrich ML
McLachlan RI (2001) Sensitive LH Histopathological Evaluation of the (2001) Testosterone inhibits
and FSH assays for monitoring low Testis. Cache River Press, spermatogonial differentiation in
serum levels in men undergoing Clearwater, FL juvenile spermatogonial depletion
steroidal contraception. Clin Ruwanpura SM, McLachlan RI, mice. Endocrinology 142:27892795
Endocrinol (Oxf) 55:331339 Meachem SJ (2010) Hormonal Shupe J, Cheng J, Puri P, Kostereva N,
Robertson KM, ODonnell L, Jones regulation of male germ cell Walker WH (2011) Regulation of
ME, Meachem SJ, Boon WC, Fisher development. J Endocrinol Sertoli-germ cell adhesion and
CR, Graves KH, McLachlan RI, 205:117131 sperm release by FSH and

151
Chapter 6: The role of testosterone in spermatogenesis

nonclassical testosterone signaling. hypogonadism: evidence of defects Vicari E, Mongioi A, Calogero AE,
Mol Endocrinol 25:238252 in the hypothalamus, pituitary, and Moncada ML, Sidoti G, Polosa P,
Singh J, ONeill C, Handelsman DJ testes. J Clin Endocrinol Metab DAgata R (1992) Therapy with
(1995) Induction of 95:30193027 human chorionic gonadotrophin
spermatogenesis by androgens in alone induces spermatogenesis in
Tan KA, De Gendt K, Atanassova N,
gonadotropin-deficient (hpg) mice. Walker M, Sharpe RM, Saunders men with isolated
Endocrinology 136:53115321 hypogonadotrophic hypogonadism
PT, Denolet E, Verhoeven G (2005)
The role of androgens in Sertoli cell long-term follow-up. Int J Androl
Sinha Hikim AP, Swerdloff RS (1999) 15:320329
Hormonal and genetic control of proliferation and functional
germ cell apoptosis in the testis. Rev maturation: studies in mice Walker WH (2009) Molecular
Reprod 4:3847 with total or Sertoli cell-selective mechanisms of testosterone action
ablation of the androgen in spermatogenesis. Steroids
Sinkevicius KW, Laine M, Lotan TL, receptor. Endocrinology 74:602607
Woloszyn K, Richburg JH, Greene 146:26742683
GL (2009) Estrogen-dependent and- Walker WH (2010) Non-classical
independent estrogen receptor- Timmons PM, Rigby PW, Poirier F actions of testosterone and
alpha signaling separately regulate (2002) The murine seminiferous spermatogenesis. Philos Trans
male fertility. Endocrinology epithelial cycle is pre-figured in the R Soc Lond B Biol Sci
150:28982905 Sertoli cells of the embryonic testis. 365:15571569
Development 129:635647 Walker WH, Cheng J (2005) FSH
Sjogren K, Liu JL, Blad K, Skrtic S,
Vidal O, Wallenius V, LeRoith D, Tsai MY, Yeh SD, Wang RS, Yeh S, and testosterone signaling in
Tornell J, Isaksson OG, Jansson JO, Zhang C, Lin HY, Tzeng CR, Chang Sertoli cells. Reproduction
Ohlsson C (1999) Liver-derived C (2006) Differential effects of 130:1528
insulin-like growth factor I (IGF-I) spermatogenesis and fertility in Wang RS, Yeh S, Tzeng CR, Chang C
is the principal source of IGF-I in mice lacking androgen receptor in (2009) Androgen receptor roles in
blood but is not required for individual testis cells. Proc Natl spermatogenesis and fertility:
postnatal body growth in mice. Proc Acad Sci USA 103:1897518980 lessons from testicular cell-specific
Natl Acad Sci USA 96:70887092 van Alphen MM, van de Kant HJ, de androgen receptor knockout mice.
Spiteri-Grech J, Weinbauer GF, Bolze Rooij DG (1988) Repopulation of Endocr Rev 30:119132
P, Chandolia RK, Bartlett JM, the seminiferous epithelium of the Weinbauer GF, Nieschlag E (1996) The
Nieschlag E (1993) Effects of FSH rhesus monkey after X irradiation. Leydig cell as a target for male
and testosterone on intratesticular Radiat Res 113:487500 contraception. In: Payne AH, Hardy
insulin-like growth factor-I and van Alphen MM, van de Kant HJ, de MP, Russell LD (eds). The Leydig
specific germ cell populations in Rooij DG (1989) Protection from Cell. Cache River Press, Vienna, IL,
rats treated with gonadotrophin- radiation-induced damage of pp 629662
releasing hormone antagonist. spermatogenesis in the rhesus
J Endocrinol 137:8189 Weinbauer GF, Behre HM, Fingscheidt
monkey (Macaca mulatta) by U, Nieschlag E (1991) Human
Suarez-Quian CA, Martinez-Garcia F, follicle-stimulating hormone. follicle-stimulating hormone exerts
Nistal M, Regadera J (1999) Cancer Res 49:533536 a stimulatory effect on
Androgen receptor distribution in Van Roijen JH, Van Assen S, Van Der spermatogenesis, testicular size, and
adult human testis. J Clin Endocrinol Kwast TH, De Rooij DG, Boersma serum inhibin levels in the
Metab 84:350358 WJ, Vreeburg JT, Weber RF (1995) gonadotropin-releasing hormone
Sun YT, Wreford NG, Robertson DM, Androgen receptor antagonist-treated nonhuman
de Kretser DM (1990) Quantitative immunoexpression in the testes of primate (Macaca fascicularis).
cytological studies of subfertile men. J Androl 16:510516 Endocrinology 129:18311839
spermatogenesis in intact and Veldhuis JD, Iranmanesh A, Samojlik Weinbauer GF, Aslam H,
hypophysectomized rats: E, Urban RJ (1997) Differential sex Krishnamurthy H, Brinkworth MH,
identification of androgen- steroid negative feedback regulation Einspanier A, Hodges JK (2001)
dependent stages. Endocrinology of pulsatile follicle-stimulating Quantitative analysis of
127:12151223 hormone secretion in healthy older spermatogenesis and apoptosis in
Sykiotis GP, Hoang XH, Avbelj M, men: deconvolution analysis and the common marmoset (Callithrix
Hayes FJ, Thambundit A, Dwyer A, steady-state sex-steroid hormone jacchus) reveals high rates of
Au M, Plummer L, Crowley WF Jr, infusions in frequently sampled spermatogonial turnover and high
Pitteloud N (2010) Congenital healthy older individuals. J Clin spermatogenic efficiency. Biol
idiopathic hypogonadotropic Endocrinol Metab 82:12481254 Reprod 64:120126

152
Chapter 6: The role of testosterone in spermatogenesis

Welsh M, Saunders PT, Atanassova N, Analysis of the testicular constitutive production of testicular
Sharpe RM, Smith LB (2009) phenotype of the Follicle- testosterone is sufficient to maintain
Androgen action via testicular Stimulating Hormone b-subunit spermatogenesis. Proc Natl Acad Sci
peritubular myoid cells is essential knockout and the activin type II USA 100:1369213697
for male fertility. FASEB J 23: receptor knockout mice by Zhao M, Baker SD, Yan X, Zhao Y,
42184230 stereological analysis. Endocrinology Wright WW, Zirkin BR, Jarow JP
Welsh M, Sharpe RM, Moffat L, 142:29162920 (2004) Simultaneous determination
Atanassova N, Saunders PT, Zhang FP, Rannikko AS, Manna PR, of steroid composition of human
Kilter S, Bergh A, Smith LB (2010) Fraser HM, Huhtaniemi IT (1997) testicular fluid using liquid
Androgen action via testicular Cloning and functional chromatography tandem
arteriole smooth muscle cells is expression of the luteinizing mass spectrometry. Steroids
important for Leydig cell function, hormone receptor complementary 69:721726
vasomotion and testicular fluid deoxyribonucleic acid from the Zhengwei Y, Wreford NG, Royce P, de
dynamics. PLoS One 5:e13632 marmoset monkey testis: absence of Kretser DM, McLachlan RI (1998)
Wilson JD (1992) Syndromes of sequences encoding exon 10 Stereological evaluation of human
androgen resistance. Biol Reprod in other species. Endocrinology spermatogenesis after suppression
46:168173 138:24812490 by testosterone treatment:
Wilson JD, Leihy MW, Shaw G, Zhang FP, Kero J, Huhtaniemi I (1998) heterogeneous pattern of
Renfree MB (2003) Unsolved The unique exon 10 of the human spermatogenic impairment. J Clin
problems in male physiology: luteinizing hormone receptor is Endocrinol Metab 83:12841291
studies in a marsupial. Mol Cell necessary for expression of the Zhou ZX, Lane MV, Kemppainen JA,
Endocrinol 211:3336 receptor protein at the plasma French FS, Wilson EM (1995)
Wistuba J, Schrod A, Greve B, Hodges membrane in the human luteinizing Specificity of ligand-dependent
JK, Aslam H, Weinbauer GF, hormone receptor, but deleterious androgen receptor stabilization:
Luetjens CM (2003) Organization of when inserted into the human receptor domain interactions
seminiferous epithelium in follicle-stimulating hormone influence ligand dissociation and
primates: relationship to receptor. Mol Cell Endocrinol receptor stability. Mol Endocrinol
spermatogenic efficiency, 142:165174 9:208218
phylogeny, and mating system. Zhang FP, Poutanen M, Wilbertz J, Zirkin BR (1998) Spermatogenesis: its
Biol Reprod 69:582591 Huhtaniemi I (2001) Normal regulation by testosterone and FSH.
Wreford NG (1995) Theory and prenatal but arrested postnatal Semin Cell Dev Biol 9:417421
practice of stereological techniques sexual development of luteinizing
Zirkin BR, Santulli R, Awoniyi CA,
applied to the estimation of cell hormone receptor knockout
Ewing LL (1989) Maintenance of
number and nuclear volume in the (LuRKO) mice. Mol Endocrinol
advanced spermatogenic cells in the
testis. Microsc Res Tech 32: 15:172183
adult rat testis: quantitative
423436 Zhang FP, Pakarainen T, Poutanen M, relationship to testosterone
Wreford NG, Rajendra Kumar T, Toppari J, Huhtaniemi I (2003) The concentration within the testis.
Matzuk MM, de Kretser DM (2001) low gonadotropin-independent Endocrinology 124:30433049

153
Chapter
Androgens and hair: a biological paradox

7 with clinical consequences


Valerie Anne Randall

7.1 Introduction 154 7.5 Androgen-dependent hair growth


7.2 Structure and function of the hair follicle 156 conditions 164
7.2.1 The roles of human hair 156 7.5.1 Androgenetic alopecia in men 164
7.2.2 Structure of the hair follicle 157 7.5.1.1 Incidence, role and
7.2.3 Changing the hair produced by a follicle consequences 164
via the hair growth cycle 158 7.5.1.2 Roles of androgens and
genes 165
7.3 The paradoxical effects of androgens on
7.5.2 Androgenetic alopecia in women 166
human hair growth 159
7.5.3 Hirsutism 166
7.3.1 Human hair growth before and after
puberty 159 7.6 Treatment of androgen-potentiated hair
disorders 167
7.3.2 Evidence for the role of androgens 160
7.6.1 Androgenetic alopecia 167
7.4 The mechanism of androgen action in the hair
7.6.1.1 Surgery 167
follicle 161
7.6.1.2 Non-hormonal therapy 167
7.4.1 Hair growth in androgen insufficiency
7.6.1.3 Endocrine-based
syndromes 161
treatments 168
7.4.2 The current model for androgen action
7.6.2 Hirsutism 168
in the hair follicle 161
7.6.2.1 Non-hormonal approaches 168
7.4.2.1 The role of the dermal papilla 161
7.6.2.2 Endocrine-based treatments 168
7.4.2.2 Paracrine factors implicated in
7.7 Key messages 169
mesenchymeepithelial interactions
in the hair follicle 163 7.8 References 170

7.1 Introduction example, the widespread customs of daily shaving of


Hair growth plays important roles in human social and mens beards and womens axillary hair in Northern
sexual communication. People throughout the world Europe and the USA. Therefore, we should not be
classify a persons state of health, sex, sexual maturity surprised that hair growth abnormalities, either more
and age, often subconsciously, by assessing their scalp or less than normal, even common male pattern
and body hair. Hairs importance is seen in many social baldness, cause widespread psychological distress.
customs in different cultures. Hair removal generally Androgens are the most obvious regulators of
has a strong depersonalizing effect, such as shaving the human hair growth. Although hair with a major
heads of soldiers, prisoners and Buddhist or Christian protective role, such as the eyelashes, eyebrows and
monks. In contrast, long hair often has positive conno- scalp hair, is produced by children in the absence of
tations, like Samsons strength in the Bible and the androgens, the formation of long pigmented hair on
uncut hair of Sikhs. Body hair is also involved; for the axillae, pubis, face etc. needs androgens in both

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

154
Chapter 7: Androgens and hair

(A) Hair changes during the human life cycle

Baby Childhood Puberty Reproductive Stages Aging Death

(B) Abnormal hair growth

Androgen insufficiency
syndromes

XY XY Female androgenetic
no androgen 5 -reductase Hirsutism alopecia
receptors deficiency
Gradual thinning on
Child hair Female hair Male hair vertex retention of
pattern only distribution distribution frontal hairline
Fig. 7.1 Human hair varies with life stage and endocrine state. (A) Changes in hair distribution and color signal a persons age, state of
maturity and sex. Visible (i.e. terminal) hair with protective functions normally develops in children on the scalp, eyelashes and eyebrows. Once
puberty occurs, more terminal hair develops on the axilla and pubis in both sexes, and on the face, chest, limbs and often back in men.
Androgens also stimulate hair loss from the scalp in men with the appropriate genes in a patterned manner, causing androgenetic alopecia. As
age increases hair follicles lose their ability to make pigment, causing graying (canities). (B) People with various androgen insufficiency
syndromes demonstrate that none of this occurs without functional androgen receptors and that only axillary and female pattern of lower
pubic triangle hairs are formed in the absence of 5a-reductase type 2. Male pattern hair growth (hirsutism) occurs in women with
abnormalities of plasma androgens or from idiopathic causes, and women may also develop a different form of hair loss, female androgenetic
alopecia or female pattern hair loss (FPHL). See plate section for color version.

sexes. In contrast, androgens may also inhibit hair sometimes stimulate hair growth, and the recognition
growth on the scalp, causing baldness (Figs. 7.1 and of its regenerative capacity. However, we still know
7.2). How one type of hormone can simultaneously relatively little about the precise functioning of this
cause these contradictory effects in the same tissue in complex cell biological system. On the other hand,
different body sites within one person is a unique our increased understanding of how androgens work
endocrinological paradox. The hair follicle has in the follicle has enabled the treatment of female
another exciting characteristic. It is the only tissue hirsutism with antiandrogens, such as cyproterone
in the adult body which can regenerate itself, often acetate, and the 5a-reductase type 2 inhibitor,
producing a new hair with different features. This is finasteride, developed to regulate prostate disorders,
how androgens can cause such major changes. is now available in many countries for use in male
Over the last 20 years, there has been much inter- pattern baldness. Greater understanding of hair fol-
est in the hair follicle, promoted by the discovery that licle biology may also enable the development of
the antihypertensive drug, minoxidil, could further approaches to treatment in the future.

155
Chapter 7: Androgens and hair

Long, thick Fig. 7.2 Androgens have paradoxically


(A) BEARD pigmented hair different effects on human hair follicles
Short, fine depending on their body site. (A) During
unpigmented and after puberty, androgens stimulate
hair the gradual transformation of small
+ follicles producing tiny, virtually colorless,
ANDROGENS vellus hairs, to terminal follicles producing
longer, thicker and more pigmented
hairs. These changes involve passing
through the hair cycle (see Fig. 7.3). (B) At
the same time, many follicles on the
scalp and eyelashes continue to produce
Vellus Terminal the same type of hairs, apparently
follicle follicle unaffected by androgens. (C) In complete
contrast, androgens may inhibit follicles
(B) NON-BALDING SCALP on specific areas of the scalp in
Androgen genetically susceptible individuals,
independent causing the reverse transformation of
terminal follicles to vellus ones, and
Long, thick
pigmented hair
androgenetic alopecia. (Diagram
reproduced from Randall 2000a.)
See plate section for color version.

+
ANDROGENS

blood
vessels

(C) BALDING SCALP


Androgen
sensitive
Long, thick
pigmented hair
Short, fine
unpigmented hair

+
ANDROGENS

Terminal Vellus
follicle follicle

People have been intrigued by the changes in


hair growth during a persons life (see Fig. 7.1) for 7.2 Structure and function of the
thousands of years, with Aristotle first recognizing hair follicle
the connection between beard growth and the testes
(reviewed by Randall 2003). This chapter will cover 7.2.1 The roles of human hair
our current knowledge of the structure and function Hairs cover almost the entire body surface of human
of hair follicles, their responses to androgens, beings except for the soles of the feet, palms of the
the mechanism of action of androgens in the hands and the lips. They are fully keratinized tubes of
follicle, and current modes of controlling androgen- dead epithelial cells where they project outside the
potentiated hair disorders. skin. They taper to a point, but otherwise are

156
Chapter 7: Androgens and hair

BALDING SCALP: androgen sensitive

+ ANDROGENS + ANDROGENS

LONG, THICK
PIGMENTED
HAIR
New New
hair hair

Club Club
hair Sebaceous hair Sebaceous
gland gland
Hair Hair
germ Original germ Original
New New
hair Dermal hair
hair hair
Dermal papilla
papilla Dermal cells
cells papilla
Dermal
Dermal papilla
papilla
TERMINAL
FOLLICLE

Fig. 7.3 Androgen inhibition of hair size on the scalp leads to balding. Hair follicles pass through regular cycles of growth (anagen), regression
(catagen), rest (telogen) and hair shedding during which the lower part of the follicle is regenerated. This enables the follicle to produce a
different type of hair in response to hormonal stimuli to coordinate to changes in the bodys development, e.g. sexual maturity or seasonal
climate changes. Androgens inhibit follicles on specific areas of the scalp in genetically susceptible individuals, causing the regenerated follicles
to be smaller, protrude less into the dermis and produce smaller, less-pigmented hairs. Eventually the terminal hairs of childhood and early
adulthood are replaced by the vellus hairs of androgenetic alopecia, and the area appears bald. Follicles must pass through a full hair cycle,
probably a succession of cycles, to accomplish major changes. (Diagram reproduced from Randall 2010.) See plate section for color version.

extremely variable in length, thickness, color and Eyelashes and eyebrow hairs prevent substances
cross-sectional shape. These differences occur entering the eyes, and head hair protects the scalp
between individuals, e.g. blonde, red or dark haired and back of the neck from sun damage during our
people, and between specific body areas within one upright posture. During puberty the development of
individual, such as the long, thick scalp and beard axillary and pubic hair signals the beginning of sexual
hairs and the short, fine ones on the back of the hand. maturity in both sexes (Marshall and Tanner 1969;
Changes also occur in some areas within an individ- Winter and Faiman 1972; 1973), while a mans beard
ual at different stages of their life; e.g. darker, thicker readily distinguishes the sexes (Jansen and Van Baalen
and longer beard hairs replace the fine, short, almost 2006), like the mane of the lion (Fig. 7.1).
colorless hairs on a boys face after puberty (Fig. 7.1).
The main functions of mammalian hair are insula- 7.2.2 Structure of the hair follicle
tion and camouflage. These are no longer necessary for Each hair is produced by a hair follicle; the biology of
the naked ape, although vestiges remain in the sea- the hair follicle was reviewed recently by Randall and
sonal patterns of our hair growth (Randall and Ebling Botchkareva (2009). Hair follicles are cylindrical, epi-
1991) and the erection of our body hairs when thelial down-growths from the epidermis into the
shivering with cold (goosebumps). Mammals often dermis and subcutaneous fat, which enlarge at the
have hairs specialized as neuroreceptors, e.g. whiskers, base into a hair bulb surrounding the tear-shaped,
and this remains, slightly, in human body hair, with its mesenchyme-derived dermal papilla (Fig. 7.3). The
good nerve supply. However, the main functions of dermal papilla, containing specialized fibroblast-like
human hair are protection and communication. cells in an extracellular matrix, regulates many aspects

157
Chapter 7: Androgens and hair

of hair growth (Reynolds and Jahoda 2004). Hairs are Hair follicle regeneration is characterized by dra-
produced by epithelial cell division in the hair bulb; matic changes in microanatomy and cellular activity
the keratinocytes move upwards, differentiating into which are still not fully understood. Hair follicle tran-
the various layers of the follicle. The central portion sition between the hair cycle stages is governed by
forms the hair itself, the color of which is produced by epithelialmesenchymal interactions between the fol-
pigment donated by the follicular melanocytes in the licular keratinocytes and the dermal papilla cells. Cell
hair bulb. By the time it reaches the surface the cells fate during hair follicle growth and involution is
are fully keratinized and dead. Each hair is sur- controlled by numerous growth regulators. During
rounded by two multi-layered epithelial sheaths: active growth and hair production, factors promoting
the inner root sheath, which helps it move through proliferation, differentiation and survival predomin-
the skin and which disintegrates when level with the ate, including WNT5a, SCF, HGF, FGF7 and IGF-1,
sebaceous gland, and the outer root sheath, which while hair follicle regression is characterized by acti-
becomes continuous with the epidermis, completing vation of various signaling pathways that induce
the skins protective barrier (Fig. 7.3). Cell division apoptosis (Botchkareva and Kishimoto 2003; Botch-
continues until the hair reaches the appropriate kareva et al. 2006).
length for its body site. The epithelial stem cells necessary to synthesize a
new hair have been identified in the bulge region of
7.2.3 Changing the hair produced the outer root sheath below the sebaceous gland (Cot-
sarelis et al. 1990; Hsu et al. 2011). The bulge contains
by a follicle via the hair growth cycle stem cells with a wide potency, which are able to
To fulfill all its roles, the type of hair produced by a replace cells of the epidermis and sebaceous glands
follicle often needs to change. Follicles possess a as well as the hair follicle (Waters et al. 2007; Shimo-
unique mechanism for this: the hair growth cycle mura and Christiano 2010), and is also associated
(Kligman 1959; Fig. 7.3). This involves destruction with the melanocyte stem cells (Nishimura 2011)
of the original lower follicle and its regeneration to and nestin-expressing stem cells which can produce
form another which can produce a hair with different nerve cell lineages (Lui et al. 2011). Recently Rabbani
characteristics. Thus postnatal follicles retain the abil- et al. (2011) have shown coordinated activation of
ity to recapitulate the later stages of follicular epithelial and melanocyte stem cells in the bulge.
embryogenesis throughout life, a unique characteris- Our understanding of this aspect of hair follicle activ-
tic in adults. Exactly how differently sized a hair can ity has expanded dramatically and is the focus of
be to its immediate predecessor is currently unclear much attention with the aim of developing the hair
because many changes, like producing a full beard follicle as a stem cell source for regenerative medicine.
(Hamilton 1958) or androgenetic alopecia The processes of the hair growth cycle allow the
(Hamilton 1951a), take several years. follicle to replace the hair with a new one which may
Hairs are produced in anagen, the growth phase. resemble the original or may be larger, smaller and/or
Once a hair reaches its full length, a short apoptosis- a different color depending on the environment or
driven involution phase, catagen, occurs, where cell stage of a mammals maturity (Fig. 7.3). Changes in
division and pigmentation stops, and the hair hair length involve alterations in the length of anagen.
becomes fully keratinized with a swollen club end This can range from two to three years or more on the
and moves up in the skin with the regressed dermal head, to produce long scalp hair (Kligman 1959), to
papilla. After a period of rest, telogen, the dermal only about two months for the short hairs on the
papilla cells and associated keratinocyte stem cells finger (Saitoh and Sakamoto 1970). These changes
reactivate, and a new lower follicle develops down- are coordinated by the pineal-hypophysis-pituitary
wards, guided by the dermal sheath which sur- system (Ebling et al. 1991; Randall 2007). Coordin-
rounded the previous follicle. The new hair then ation to the environment is particularly important for
grows up into the original upper follicle (Fig. 7.3). some mammals, such as mountain hares, which need
The existing hair is generally lost; although previously a longer, warmer and white coat in the snowy winter,
believed due to the new hairs upward movement, a but a shorter, brown coat in the summer to increase
further active shedding stage, exogen, is now recog- their chances of survival (Flux 1970). Human beings
nized (Higgins et al. 2009). from temperate regions also exhibit seasonal changes

158
Chapter 7: Androgens and hair

in both scalp (Orentreich 1969; Randall and Ebling such as Hairless and Tabby, plus transmembrane
1991; Courtois et al. 1996) and body hair (Randall molecules and extracellular matrix molecules have
and Ebling 1991), in line with seasonal changes in all been implicated in the mesenchymalepithelial
melatonin, prolactin and cortisol secretion (Wehr interactions (Rendl et al. 2005).
1998; Wehr et al. 2001). The main change in human One of the first signs of puberty is the gradual
hair growth is the production of adult patterns of appearance of a few larger and more pigmented inter-
body hair growth after puberty, like the male lions mediate hairs, first in the pubic region and later in the
mane, in response to androgen (Fig. 7.2); some sea- axillae. Intermediate-sized hair follicles have recently
sonal fluctuations in human body hair growth may been characterized on the face (Miranda et al. 2010).
also coordinate at least in part to those of androgens These are replaced by longer and darker terminal
(Randall and Ebling 1991; Randall 2008). hairs (Fig. 7.2), and the area spreads. In boys, similar
These annual seasonal changes are important for changes occur gradually on the face starting above the
any investigations of androgen-dependent or scalp mouth and on the central chin, eventually generally
hair growth, particularly in individuals living in tem- spreading over the lower part of the face and parts of
perate zones. For example, scalp hair loss may be the neck, readily distinguishing the adult male (Mar-
exacerbated during the increased autumnal shedding shall and Tanner 1969; 1970). Adult mens pubic hair
in both male and female patients. This has particu- distribution also differs from womens, extending in a
larly important implications for any assessments of diamond shape up to the navel in contrast to womens
new therapies or treatments to stimulate, inhibit or inverted triangle. Terminal hair on the chest and
remove hair; to be accurate, measurements need to be sometimes the back is also normally restricted to
carried out over a year to avoid natural seasonal men, though both sexes may also develop intermedi-
variations (Randall 2008). ate terminal hairs on their arms and legs, with ter-
minal hairs normally restricted to the lower limbs in
7.3 The paradoxical effects of women (Fig. 7.1). In all areas the responses are grad-
ual, often taking many years. Beard weight increases
androgens on human hair growth dramatically during puberty, but continues to rise
7.3.1 Human hair growth before until the mid-thirties (Hamilton 1958); while terminal
hair growth on the chest and in the external ear canal
and after puberty may be first seen many years after puberty (Hamilton
In utero the human body is covered with quite long, 1946).
colorless lanugo hairs. These are shed before birth and The amount of body hair is very variable and
at birth, or shortly after; babies normally exhibit differs both between families within one race and
pigmented, quite thick protective hairs on the eye- between races, with Caucasians generally exhibiting
brows and eyelashes and variable amounts on the more than Japanese (Hamilton 1958). This implicates
scalp, and by the age of three or four the scalp hair a genetically determined response to circulating trig-
is usually quite well developed, though it will not yet gers. The responses of the follicles themselves also
have reached its maximum length. These readily vis- vary, with female hormone levels being sufficient to
ible pigmented hairs are known as terminal hairs and stimulate terminal hair growth in the pubis and axil-
are formed by large deep terminal follicles (Fig. 7.2). lae, but male hormones being required for other
This emphasizes that terminal hair growth on the areas, such as the beard and chest. Beard hair growth
scalp, eyelashes and eyebrows is not androgen also remains high, even after 70, while axillary growth
dependent. The hairless rest of the body is normally is maximal in the mid-twenties and falls quite rapidly
covered with fine, short almost colorless vellus hairs then in both sexes (Hamilton 1958). This also seems
produced by small, short vellus follicles (Fig. 7.2). The paradoxical; markedly different responses in the two
molecular mechanisms involved in the distribution areas, although both respond to apparently similar
and formation of the different types of follicles during stimulation by androgens.
embryogenesis are not clear, but secreted signaling During early puberty the frontal hair line is
factors such as Sonic hedgehog, WNT and growth usually straight across the top of the forehead.
factors (e.g. the EGF and FGF families), nuclear With increasing age there is frequently a progressive
factors including various homeobox genes and others regression of the frontal hair line in a prescribed

159
Chapter 7: Androgens and hair

(A)

I II IIa III IIIa III-vertex

IV IVa V Va VI VII

(B)

Stage I II III
Fig. 7.4 Patterns of hair loss in androgenetic alopecia in men and women differ. Androgens cause a gradual inhibition of hair growth on the
scalp in genetically predisposed individuals. This is much more common in men than in women, and the pattern of the hair loss in men (A;
Hamilton 1951a) differs from that in women (B; Ludwig 1977). In men, the first signs are generally temporal regression, which spreads
backwards and joins thinning regions on the vertex to give a bald crown. In women, the front hairline is normally retained, and a general
thinning on the vertex gradually becomes more pronounced until the vertex becomes bald. It is also less clear whether androgens are actually
involved in women.

manner (described below in Section 7.5.1), accom- retained when follicles are transplanted to other skin
panied by progressive thinning of terminal hair on sites (Ebling and Johnson 1959), the basis of correct-
the vertex (Fig. 7.4). This is characterized by a gradual ive hair follicle transplant surgery (Orentreich and
inhibition of terminal follicles to smaller vellus fol- Durr 1982; Gokrem et al. 2008).
licles (Fig. 7.3), with the length of anagen decreasing
and that of telogen increasing. This is an example of a
much more dramatic biological paradox. How does 7.3.2 Evidence for the role of androgens
one hormone stimulate hair growth in many areas Although androgens are the clearest regulators of
such as the face, have no effect in others, e.g. eye- human hair growth unlike most other mammals
lashes, while inhibiting follicles on the scalp? These (Ebling et al. 1991), various other circulating factors
contrasts are presumably due to differential gene have an effect (reviewed in Randall 2007). These
expression within follicles from the various body sites. include adequate nutritional supplies, due to the fol-
The intrinsic response of individual follicles is licles high metabolic demands (Bradfield 1971), the

160
Chapter 7: Androgens and hair

hormones of pregnancy, which cause a prolonged ana- and female pattern pubic hair, but very little beard
gen resulting in a synchronized shedding of a propor- growth; they are also not reported to show male
tion of scalp hairs post-partum (Lynfield 1960), and pattern baldness (Griffin and Wilson 1989;
lack of thyroid hormone which restricts hair growth Fig. 7.3). Although the identification of two forms
(Jackson et al. 1972). Growth hormone is also neces- of 5a-reductase (type 1 and type 2) makes the situ-
sary in combination with androgens for normal body ation complex, all individuals with 5a-reductase
hair development in boys (Zachmann and Prader deficiency so far are deficient in 5a-reductase type 2,
1970; Zachmann et al. 1976). There is much evidence confirming its importance for much androgen-
supporting androgens importance, which fits in well dependent hair growth. A role for 5a-reductase in
with the concept of much terminal hair growth being a baldness is also supported by the ability of oral
secondary sexual characteristic. Terminal hair appear- finasteride, a 5a-reductase type 2 inhibitor (Kaufman
ance in puberty parallels the rise in circulating andro- et al. 1998; Shapiro and Kaufman 2003) and the dual
gen levels and occurs later in boys than girls (Marshall 5a-reductase inhibitor, dutasteride (Olsen et al.
and Tanner 1969; 1970; Winter and Faiman 1972; 2006), to promote hair regrowth. This suggests that
1973). Testosterone also stimulates beard growth in the formation of terminal pubic and axillary hair can
eunuchs and elderly men (Chieffi 1949). An extensive be mediated by testosterone itself, while that of the
US study also showed that castration before puberty secondary sexual hair of men requires the presence
prevented beard and axillary hair growth and after of 5a-dihydrotestosterone. This demonstrates a third
puberty reduced them (Hamilton 1951b; 1958). Never- paradox in androgen effects on hair follicles. Why
theless, the strongest evidence for the essential nature does the stimulation of increasing size in some
of androgens is the lack of any body hair, even the follicles like beard require 5a-dihydrotestosterone
female pubic and axillary pattern, or evidence of any formation, while follicles in the axillary and pubic
male pattern baldness, in adult XY androgen insensi- regions carry out the same changes in the absence of
tivity patients with absent or dysfunctional androgen 5a-dihydrotestosterone? Since androgens are stimu-
receptors, despite normal or raised circulating levels of lating the same transformation, presumably via the
androgens (Fig. 7.1, B) (see Chapter 3). same receptor, this is currently difficult to under-
stand, although it is further evidence of the intrinsic
7.4 The mechanism of androgen differences within hair follicles. It suggests that some
less well-known aspect of androgen action is
action in the hair follicle involved in hair follicles normally specific to men,
7.4.1 Hair growth in androgen which requires 5a-dihydrotestosterone, such as inter-
action with a specific transcription factor. Interest-
insufficiency syndromes ingly, androgen-dependent sebum production by
As described in Chapter 2 of this book, androgens the sebaceous glands attached to hair follicles is also
from the blood stream enter the cell and bind to normal in 5a-reductase deficiency type 2 (Imperato-
specific, intracellular androgen receptors, usually in McGinley et al. 1993).
the form of testosterone or its more potent metabol-
ite, 5a-dihydrotestosterone. The hormone-receptor
complex, generally in combination with transcrip- 7.4.2 The current model for androgen
tional regulators, then activates the appropriate gene
transcription for that cell type. Androgen insuffi- action in the hair follicle
ciency patients without functional androgen receptors 7.4.2.1 The role of the dermal papilla
demonstrate the absolute requirement for androgen The mesenchyme-derived dermal papilla plays a
receptors within hair follicles for the development of major role in determining the type of hair produced
the hair growth ascribed in Section 7.3.2 to androgens by a follicle, as shown by an elegant series of experi-
(see Chapter 3). These individuals produce no body ments involving the rat whisker by Oliver, Jahoda,
hair at puberty, even with high circulating androgen Reynolds and colleagues (Reynolds and Jahoda
levels, nor do they go bald (Fig. 7.1). 2004). Whisker dermal papillae transplanted into ear
Men with 5a-reductase deficiency also contribute or glabrous skin initiated the production of whisker
to our understanding because they exhibit axillary follicles, and hair growth could also be stimulated by

161
Chapter 7: Androgens and hair

Fig. 7.5 Current model of androgen


Hair action in the hair follicle: in the current,
well-accepted hypothesis, androgens
Hair bulb from the blood enter the hair follicle via
the dermal papillas blood supply. If
Basement membrane Melanocyte appropriate, they are metabolized to
(indirect target) 5a-dihydrotestosterone. They bind to
? androgen receptors in the key regulatory
? Extracellular matrix
cells of the follicle, the dermal papilla cells
Epithelial cells ? +T causing changes in their production of
(indirect target) +T ? regulatory paracrine factors; these then
+T Dermal papilla cells
(direct target)
alter the activity of dermal papilla cells,
? follicular keratinocytes and melanocytes.
? T = testosterone; ? = unknown paracrine
Blood capillary T
factors. (Modified from Randall 2000b.)
T See plate section for color version.
T
T CIRCULATING ANDROGENS T T

cultured dermal papilla cells reimplanted in vivo such as beard (Randall et al. 1992) and balding scalp
(Jahoda et al. 1984). (Hibberts et al. 1998) contain higher levels of specific,
In many embryonic steroid-regulated tissues, saturable androgen receptors than androgen-insensi-
including the prostate and the breast, steroids act via tive non-balding scalp in vitro; this has been con-
the mesenchyme (Thomson et al. 1997). Since hair firmed by studies using RT-PCR (reverse
follicles recapitulate the stages of embryogenesis transcription polymerase chain reaction) (Ando
during their growth cycles to reform a new lower hair et al. 1999). Most importantly, metabolism of testos-
follicle, they may behave like an embryonic tissue in terone by cultured dermal papilla cells also reflects
the adult. Studies on testosterone metabolism in vitro hair growth in 5a-reductase deficiency patients with
by plucked hair follicles, which leave the dermal beard, but not pubic or non-balding scalp, cells
papilla behind in the skin, from different body sites, forming 5a-dihydrotestosterone in vitro (Itami et al.
did not reflect the requirements for 5a-reductase in 1990; Thornton et al. 1993; Hamada et al. 1996);
vivo (reviewed in Randall 1994; Randall et al. 1991), similar results have been obtained examining gene
leading to the hypothesis that androgens would act on expression of 5a-reductase type 2 by RT-PCR (Ando
the other components of the hair follicle via the et al. 1999). This has led to wide acceptance of the
dermal papilla (Randall et al. 1991; Randall 2007). In hypothesis (Randall 2007; 2008).
this hypothesis androgens would alter the ability of However, some more recent observations suggest
the dermal papilla cells to synthesize, or release, con- minor modifications. The lower part of the connect-
trolling factors which would affect follicular keratino- ive tissue sheath, or dermal sheath, which surrounds
cytes, melanocytes and connective tissue sheath cells, the hair follicle and isolates it from the dermis, can
and also probably the dermal endothelial cells, to alter form a new dermal papilla and stimulate new human
the follicles blood supply in proportion to its change hair follicle development in another person (Reynolds
in size (Fig. 7.5). These factors could be growth et al. 1999). Cultured dermal sheath cells from beard
factors and/or extracellular matrix proteins. This follicles contain similar levels of androgen receptors
model would facilitate a mechanism for precise con- to beard dermal papilla cells (unpublished data: Mer-
trol during the complex changes needed to alter a rick AE, Randall VA, Messenger AG, Thornton MJ.
follicles size in response to androgens. Beard dermal sheath cells contain androgen receptors:
This hypothesis has now received a great deal of implications for future inductions of human hair
experimental support. Androgen receptors have been follicles.), and balding scalp dermal sheath expresses
localized by immunohistochemistry in the dermal the mRNA for 5a-reductase type 2 like the dermal
papilla and not the keratinocyte cells (Choudhry papilla (Asada et al. 2001), indicating that the
et al. 1992; Itami et al. 1995a). Cultured dermal dermal sheath can respond to androgens without
papilla cells derived from androgen-sensitive follicles the dermal papilla acting as an intermediary. Clearly

162
Chapter 7: Androgens and hair

the dermal sheath also plays an important role in the et al. 1995a) and keratinocytes (Hibberts and Randall
hair follicle. This may be as a reserve to replace the 1996). In contrast, testosterone decreased the mito-
key inductive and controlling role of the dermal genic capacity of androgenetic alopecia dermal papilla
papilla cells if they are lost, to ensure follicles are cells from both men (Hibberts and Randall 1996) and
not lost. Alternatively, or in addition, the dermal stump-tailed macaques (Obana et al.1997). Interest-
sheath cells may respond directly to androgens to ingly, conditioned media from balding cells inhibited
facilitate the increase or decrease in size of the sheath not only the growth of other human dermal papilla
or even the dermal papilla in the development of a cells but also mouse hair growth in vivo (Hamada and
new anagen follicle; this would enable the new hair Randall 2006). This implies that an autocrine mech-
follicle to be larger or smaller depending on the fol- anism is involved in androgen-mediated hair follicle
licles specific response to androgens. changes; a view which is supported by the actual
In addition, studies on the beard hair medulla, a changes that occur in dermal papilla cell numbers
central structure only seen in the middle of large and size (Elliott et al. 1999). As well as supporting
human hairs, showed coexpression of a specialized the hypothesis for the mechanism of action, these
keratin, hHa7, and the androgen receptor (Jave- results demonstrate that the paradoxical effects of
Suarez et al. 2004). Since the hHa7 gene promoter androgen on hair follicles observed in vivo are
also contained sequences with high homology to the reflected in vitro, strengthening the use of cultured
androgen response element (ARE), keratin hHa7 dermal papilla cells as a model system for studying
expression may be androgen regulated. However, androgen action in vitro.
no stimulation occurred when the promoter was The main emphasis of research now lies in identi-
transfected into prostate cells, and keratin hHa7, with fying specific factors whose production by dermal
the same promoter, is also expressed in androgen- papilla cells is altered by androgens (reviewed by
insensitive body hairs of chimpanzees, making the Randall 2007). To date only insulin-like growth factor
significance unclear. Nevertheless, the current model (IGF-1), a potent mitogen, is identified as androgen-
needs modification to include possible specific, direct stimulated in beard cells in vitro (Itami et al. 1995b).
action of androgens on lower dermal sheath and DNA microarray methods also revealed that three
medulla cells. genes, SFRP2, MN1, and ATP1B1, were expressed at
significantly higher levels in beard than normal scalp
cells, but no changes were detected due to androgen in
7.4.2.2 Paracrine factors implicated in mesenchyme vitro (Rutberg et al. 2006). However, stem cell factor
epithelial interactions in the hair follicle (SCF), the ligand for the melanocyte receptor c-kit, is
The production of growth factors by cultured dermal secreted in greater quantities by dermal papilla cells
papilla cells derived from human and rat hair follicles derived from beard hair follicles (with generally
has been investigated by several groups on the basis of darker hairs) than non-balding scalp cells (Hibberts
the primary role of the dermal papilla, its potential et al. 1996b), while cells from balding follicles (which
probable role in androgen action and the retention of have pale hairs) produce significantly less. However,
hair growth-promoting ability by cultured rat cells the concentration and distribution of the melanocytes
(discussed above). Cultured dermal papilla cells in the balding scalp follicles remain unchanged (Ran-
secrete both extracellular matrix factors and soluble, dall et al. 2008). Since SCF plays important roles in
proteinaceous growth factors (Randall 2007). Bio- hair pigmentation (reviewed in Randall et al. 2008),
assays demonstrate that human dermal papilla cells the dermal papilla probably provides local SCF for
secrete factors which stimulate the growth of other follicular melanocytes. Androgens in vivo appear to
dermal papilla cells (Randall et al. 1991; Thornton alter SCF expression by facial dermal papilla cells to
et al. 1998), outer root sheath cells (Itami et al. change melanocyte activity and thus hair color.
1995a), transformed epidermal keratinocytes (Hib- Androgens also inhibit balding dermal papilla
berts and Randall 1996) and endothelial cells (Hib- cells gene expression of protease nexin-1, a potent
berts et al. 1996a). Importantly, testosterone in vitro inhibitor of serine proteases which regulate cellular
stimulated greater mitogenic capacity of beard cells to growth and differentiation in many tissues (Sonoda
affect beard, but not scalp, dermal papilla cells et al. 1999). This may act by altering the amount of
(Thornton et al. 1998), outer root sheath cells (Itami extracellular matrix components produced and

163
Chapter 7: Androgens and hair

therefore the size of the follicle and hair produced androgenetic alopecia develops, anagen becomes
(Elliott et al. 1999).The inhibitory effects of media shorter, resulting in shorter hairs and an increasing
conditioned by balding dermal papilla cells on dermal percentage of hairs in telogen (see Section 7.2.3);
papilla cell growth in vitro and mouse hair growth in follicle miniaturization is also seen histologically,
vivo (Hamada and Randall 2006) suggest the active and the hairs become thinner (Braun-Falco and
secretion of an inhibitory factor(s). The recent dem- Christophers 1968; Kligman 1988; Rushton et al.
onstration that bald scalp from men with androge- 1991; Whiting 1993). The connective tissue sheath left
netic alopecia retains hair follicle stem cells but lacks in the dermis when the follicle becomes miniaturized
CD200-rich and CD34+ hair pigmentor cells also may become subject to chronic inflammation; this
suggests that inhibitory factors are involved (Garza may prevent terminal hair regrowth in long-term
et al. 2011). Androgens do stimulate the production baldness (Kligman 1988).
of transforming growth factor beta1 (TGF-b1) in Balding occurs in men in a precise pattern
balding dermal papilla cells with transfected androgen described by Hamilton (1951a), starting with regres-
receptors (Inui et al. 2002) and TGF-b2 (Hibino and sion of the frontal hairline in two wings and balding
Nishiyama 2004) in natural balding cells. Transform- in the center of the vertex. Hamilton graded this
ing growth factor beta is a strong candidate for an progression from type I, prepubertal scalp with ter-
inhibitor of keratinocyte activity in alopecia, as it minal hair on the forehead and all over the scalp,
inhibits hair follicle growth in vitro (Philpott 2000), through gradual regression of the frontal hairline
and a probable suppressor of TGF-b1 delayed catagen and thinning on the vertex, to type VII, where the
progression in mice in vivo (Tsuji et al. 2003), bald areas became fully coalesced to leave hair only
although TGF-b2 and TNF-a were actually slightly around the back and sides of the head (Fig. 7.4).
reduced in balding cells in a limited DNA macroarray Norwood modified Hamiltons classification, includ-
analysis (Midorikawa et al. 2004). In addition, 5a- ing variations for the middle grades; this scale is used
dihydrotestosterone did increase the production of extensively during clinical trials (Norwood 1975). The
dickkopf 1 (DKK1), which stimulated apoptosis in physiology and pathophysiology of androgenetic alo-
keratinocytes (Kwack et al. 2008), and inhibited ker- pecia is reviewed more fully by Randall (2000b; 2005;
atinocyte growth by modifying WNT signaling in 2010), and an approach to its diagnosis was recently
balding dermal papilla cells (Kitagawa et al. 2009). reported by the European Consensus Group (Blume-
Thus, studying dermal papilla cells already impli- Peytavi et al. 2011a).
cates several factors: IGF-1 in enlargement; SCF in
altered pigmentation; and nexin-1, TGF-b, and DKK1 7.5.1.1 Incidence, role and consequences
in miniaturization. Alterations in several factors are The incidence of androgenetic alopecia in Caucasians
probably necessary to precisely control the major cell is high, with estimates varying widely, but progression
biological rearrangements required when follicles to type II is detected in 95% of men (Hamilton 1951a).
change size. Further study of this area should increase It is also seen in other primates, being well studied in
our understanding of the complex hair follicle and the stump-tailed macaque. This suggests a natural
lead to better treatments for hair follicle disorders. progression of a secondary sexual characteristic rather
than the malfunction of a disease. Marked androge-
netic alopecia would obviously highlight the surviving
7.5 Androgen-dependent hair older man as a leader, like the silver back of the chief
growth conditions male gorilla and the larger antlers of the mature deer
stags. Others have speculated that the flushed bald skin
7.5.1 Androgenetic alopecia in men would look aggressive to an opponent (Goodhart
A generalized loss of hair follicles from the scalp 1960) or mean there was less hair for the opposition
known as senescent balding has been reported in both to pull (Ebling 1985), giving the bald man important
sexes by the seventh or eighth decade (Kligman 1988; advantages. The lower incidence of androgenetic
Courtois et al. 1995). This differs from the progressive alopecia amongst men from African races (Setty
baldness seen in androgenetic alopecia, also known as 1970) suggests that any advantages did not outweigh
male pattern baldness, male pattern alopecia, common the evolutionary survival advantages of the hairs pro-
baldness or androgen-dependent alopecia. As tection of the scalp from the hot tropical sun.

164
Chapter 7: Androgens and hair

In the current youth-orientated culture of indus- (Ellis and Harrap 2001). Interestingly, a very strong
trialized societies, the association of increasing hair correlation in incidence was found in 54 sets of sons
loss with age combined with the major role of hair in and fathers, with 81.5% of balding sons having
human communication means that androgenetic alo- balding fathers (HamiltonNorwood scale III or
pecia has strong negative connotations. It often causes higher) (Ellis and Harrap 2001; Ellis et al. 2001a).
well-documented psychological distress and reduc- This is greater than expected from an autosomal
tion in the quality of life, even though it is not life- dominant inheritance and could imply a paternally
threatening or physically painful, in both men (e.g. inherited gene, e.g. on the Y chromosome, or the
Franzoi et al. 1990; Girman et al. 1998) and women involvement of a gene that is capable of being pater-
(Van der Donk et al.1991; Cash 1993). Other people nally imprinted (i.e. preferentially inactivated by
perceive men with visible hair loss as older, less phys- methylation of DNA, etc.).
ically and socially attractive, weaker and duller. In Several genes have been investigated for associ-
parallel, people with androgenetic alopecia have a ation with androgenetic alopecia. No association was
poor self-image, feel older and are lacking in self- detected with neutral polymorphic markers for either
confidence, even those who seem accepting of their type 1 (SRD5A1) or type 2 (SRD5A2) 5a-reductase
condition and have never sought treatment (Girman genes in casecontrol association studies of Austra-
et al.1998), and this has recently been shown to apply lian (Ellis et al. 1998) or Korean (Asian) men (Ha
to men from many countries (Cash 2009). Male pat- et al. 2003). A later study found an association with a
tern baldness primarily causes concern amongst those mutant allele (A49T) of 5a-reductase type 2, but this
who develop marked loss before their forties, and decreased the incidence of alopecia, although increas-
early balding has been linked to myocardial infarction ing that of prostate cancer (Hayes et al. 2006)!
(Lesko et al. 1993). Whether this indicates a dual end- Known dimorphic and polymorphic markers within
organ sensitivity or reflects the psychological stress the androgen receptor gene are more linked to
early balding induces in the youth-orientated Ameri- balding in Caucasian men (Ellis et al. 2001b). The
can culture is unknown. No relationship between the Stu I restriction fragment length polymorphism
incidence of balding and prostatic carcinoma was (RFLP) in exon 1 was present in 98% of 54 young
detected in men aged between 50 and 70 by balding men and 92% of 392 older balding men, but
Demark-Wahnefried et al. (1997), while others report was also found in 77% of their older, non-balding
both an increased (Giles et al. 2002; Yassa et al. 2011) controls. Analysis of triplet repeat polymorphisms,
and a reduced relative risk (Wright et al. 2010). Since CAG and GAC, revealed significantly higher inci-
both conditions have very high incidence in Cauca- dence of short/short polymorphic CAG/GGC haplo-
sians, it would be unlikely that there would be a types in balding subjects and lower short/long,
specific clear-cut relationship that could be useful although no significance was provided. Interestingly,
clinically to predict cancer risk from hair loss. shorter triplet repeat lengths are associated with
precocious puberty, i.e. appearance of pubic hair
7.5.1.2 Roles of androgens and genes before age eight (Hoffmann and Happle 2000) and
Male pattern baldness is androgen dependent, since it androgen-dependent prostate cancer (Stanford et al.
does not occur in castrates, unless they are given 1997). Whether this has functional significance, such
testosterone (Hamilton 1942), nor in XY individuals as increased androgen sensitivity, or simply reflects
with androgen insensitivity due to non-functional linkage disequilibrium with a causative mutation, is
androgen receptors (see Chapter 3). The genetic not clear. However, when the binding capacity for a
involvement in androgenetic alopecia is also pro- range of steroids was compared between androgen
nounced. It runs in families, there are racial differ- receptors from balding and non-balding follicle
ences, and androgen replacement stimulated balding dermal papilla cells, no differences were detected
only in castrated men with a family history (Hamilton (Hibberts et al. 1998), and no link was seen with
1942). Although androgenetic alopecia has historic- increased copy number variations of the androgen
ally been accepted as an autosomal dominant trait receptor gene (Cobb et al. 2009).
with variable penetrance (Bergfeld 1955), this is based Recently, genetic variability in a 1 Mb region
on a familial analysis in 1916 (Osbourn 1916), and a within and centromeric to the androgen receptor gene
more complex, polygenic inheritance is more likely was found associated with androgenetic alopecia

165
Chapter 7: Androgens and hair

(Hillmer et al. 2008), and the strongest risk was abnormality (reviewed by Cousen and Messenger
associated with a variant in the flanking ectodyspla- 2010). Recent report of an XY individual with com-
sin A2 receptor gene (EDA2R) (Hillmer et al. 2009). plete androgen insensitivity and no other androgen-
Links with a locus on chromosome 20 (20 pll) have dependent characteristics who exhibits female pat-
also been reported in several populations (Hillmer tern hair loss (Cousen and Messenger 2010) does
et al. 2008; Richards et al. 2008). Other genes have suggest that sometimes at least there may be an
also been implicated, including a link to one allele of androgen-independent, or androgen receptor-inde-
the steroid metabolism gene, CYP17, to both women pendent, mechanism. Overall, there is some debate
with polycystic ovaries and their brothers with early about whether androgen is essential for this hair loss
onset androgenetic alopecia (Carey et al. 1993). An in women (Birch et al. 2002), though this is still
interesting connection is severe, early onset androge- generally assumed. If, as occurs in men, the changes
netic alopecia in men with the X-linked gene for develop due to the genetically influenced, specific
adrenoleukodystrophy, who tend to have low testos- follicular responses within the scalp follicles them-
terone levels (Konig et al. 2000). The gene for hair- selves, it is not surprising that circulating androgen
less, which results in a complete loss of hair (Ahmad abnormalities are often absent.
et al. 1998), also showed a marginally significant
correlation with androgenic alopecia with two muta-
tions, but these became insignificant after correction 7.5.3 Hirsutism
for multiple testing (Hillmer et al. 2002). The situ- Hirsutism is the development of male pattern body
ation is still not fully clear, but is moving forward hair growth in women. This normally benign con-
rapidly. dition causes marked psychological distress because
the person erroneously feels that they are changing
sex. The extent of body hair growth which causes a
7.5.2 Androgenetic alopecia in women problem varies and depends on the amount of
Androgenetic alopecia has also been described in normal body hair amongst her race or subgroup.
women, but the pattern of expression is normally Normally hirsutism would include terminal hair on
different and the incidence less. Post-pubertal reces- the face, chest or back. Ferriman and Gallwey
sion to type II was found in about 25% of Caucasian (1961) introduced a scale for grading hirsutism
women by age 50 (Hamilton 1951a), although this which is widely used, especially to monitor hirsut-
did not develop further. Although women can ism progression with, or without, treatment. An
exhibit the male pattern, they generally do not approach for the evaluation of hirsutism has
show the frontal recession and usually exhibit a recently been established as a European consensus
different pattern, retaining the frontal hair line but (Blume-Peytavi et al. 2009).
having a progressive diffuse loss or thinning on the Hirsutism is often associated with an endocrine
vertex which may lead to balding (Ludwig 1977) abnormality of the adrenal or ovary which causes
(Fig. 7.4). This type of balding is frequently termed raised androgens and is very frequently (over 70%
female pattern hair loss (FPHL) rather than andro- of cases) associated with polycystic ovarian syn-
genetic alopecia. Venning and Dawber (1988) found drome (PCOS). It can also be caused by Cushing
that 80% of premenopausal women had thinning in syndrome, acromegaly, hyperprolactinemia and
Ludwig stages IIII; while 13% had Hamilton types drugs (Azziz 2003; Alsantali and Shapiro 2009).
IIIV. After the menopause, 37% exhibited the Some women have no obvious underlying clinical
male pattern with some showing marked templar or biochemical disorder and are termed idio-
M-shaped recession, although not progressing pathic. The proportion of these is larger in older
beyond Hamilton stage IV. papers as modern methods increase the range of
The progression of balding in women is normally abnormalities that can be detected, e.g. low SHBG.
slow, and a full endocrinological investigation is The assumption that idiopathic hirsutism is due to
generally recommended if a rapid onset is seen. a greater sensitivity of the follicles to normal andro-
Although female pattern hair loss is seen frequently gens is given credence by hirsutism occurring asym-
in association with hyperandrogenism, other women metrically on only one side of a woman (Jenkins
frequently have no other symptoms of androgen and Ash 1973).

166
Chapter 7: Androgens and hair

7.6 Treatment of androgen- 25 years. However, its mechanism of action has been
very uncertain (Messenger and Rundegren 2004).
potentiated hair disorders Recent detection of appropriate ATP-sensitive potas-
7.6.1 Androgenetic alopecia sium (KATP) channels in the dermal papilla of human
hair follicles, and appropriate responses in human
Mainly because the pathogenic mechanisms of andro-
hair follicles in organ culture (Shorter et al. 2008)
genetic alopecia are not fully understood, the treat-
has confirmed a probable action via these channels.
ments available are limited and vary in effectiveness.
Minoxidil stimulates regrowth in up to 30%, with
Over the centuries, a wide range of remedies has been
only about 10% obtaining complete regrowth; most
suggested (Lambert 1961), and current treatments
success occurs with younger men and with the early
include wigs and hairpieces, surgery and medical
stages of balding, i.e. Hamilton stage V or less (Daw-
therapies including hormone action modifiers and
ber and Van Neste 1995). A stronger topical applica-
non-hormonal therapy (Ross and Shapiro 2005;
tion of a 5% solution is now licensed for use in men in
Rogers and Avram 2008). Several of these are based
many countries (Olsen et al. 2002), and recently a
on our understanding of the mechanisms of androgen
more acceptable, user-friendly, foam-based applica-
action within the follicle. Recently some investigators
tion has been developed (Blume-Peytavi 2011b).
have also started looking into the application of lasers
Recently, prostaglandin F2a (PGF2a) analogs, such
for androgenetic alopecia based on the low levels of
as latanoprost and bimatoprost, used as eye drops to
visible or near infrared light used to reduce inflam-
reduce intraocular pressure in glaucoma, have been
mation etc. (Leavitt et al. 2009; Kim et al. 2011).
reported to stimulate eyelashes to lengthen, thicken
and darken (Curran 2009). Bimatoprost (Latisse), a
7.6.1.1 Surgery prostamide F2a analog, was licensed for the treatment
All surgical methods capitalize on the different intrin- of hypotrichosis of the eyelashes by the FDA in
sic responses to androgens by spreading non- December 2008 (NDA 022369). No other drugs have
balding, i.e. occipital and parietal, terminal follicles caused such a dramatic stimulation of hair growth
over the androgen-sensitive scalp regions (Orentreich after a local application as these prostaglandin analogs
and Durr 1982). Originally involving the transplant of have on eyelashes; minoxidils effect topically is much
small biopsies with several follicles, this usually now less than via the original oral ingestion (Messenger
involves micro-grafts with one or two follicles. Once and Rundegren 2004). The eyelashes are highly spe-
established, these expensive and painful treatments cialized, short hairs which have evolved to protect the
are long-lasting; however, the effect can be marred eyes from foreign objects. They have marked differ-
by the continual natural progression of balding, ences to scalp hairs, and their follicles lack the normal
which may well require further transplants to avoid arrector pili muscle found in scalp follicles (Thibaut
isolation of the transplanted region. Future modifica- et al. 2009). Although specialized, eyelashes are pro-
tions may include culturing dermal papilla cells to duced by hair follicles and are replaced regularly via
expand the non-balding follicular material before the hair cycle (Thibaut et al. 2009), like all other hair
replanting into balding regions (Randall 2010; Aoi follicles. However, human hair follicles also exhibit
et al. 2012). markedly different behaviors depending on their body
site, including the major differences in response to
7.6.1.2 Non-hormonal therapy androgens discussed above, and hair graying with age
The ideal treatment for alopecia is an externally occurs first above the ears before gradually spreading
applied (topical) substance which would act locally over the scalp (Keogh and Walsh 1965). We have
to stimulate hair growth only in specific target areas, carried out a range of studies on scalp hair follicles
with no side-effects. Minoxidil, a vasodilator used for and shown that bimatoprost can stimulate hair follicle
hypertension, stimulated excessive hair growth as a growth in organ culture and that scalp follicles con-
side-effect. This provoked major interest in hair fol- tain appropriate receptors to respond in vivo (unpub-
licle biology because it demonstrated that vellus fol- lished data: Khidhir KG, Woodward DF, Farjo NP,
licles could be stimulated to form terminal hairs. Farjo BK, Tang ES, Wang JW, Picksley SM, Randall
Topical application of minoxidil has been used in VA. A prostaglandin-related glaucoma therapy offers
both male and female androgenetic alopecia for over a novel approach for treating alopecia). Based on this,

167
Chapter 7: Androgens and hair

clinical trials of bimatoprost applied topically in showed similar, possibly better effects than finasteride
both men and women are now beginning in the in men (Olsen et al. 2006).
USA and Germany. A short study of another glau- Although a range of treatments is now available,
coma drug, latanoprost, a PGF2a analog, has reported all medications need to be used continually because
some effects on androgenetic alopecia in men they are opposing a natural process which, if treat-
(Blume-Peytavi et al. 2011c). ment is discontinued, retains all the components to
continue to progress.
7.6.1.3 Endocrine-based treatments
Antiandrogen therapy is not a practical option for
men due to the side-effects, but cyproterone acetate,
7.6.2 Hirsutism
in combination with estrogen to ensure contraception Once a serious underlying pathology has been elimin-
and prevent potential feminization of a male fetus, ated, a range of treatments is available for hirsutism
has been used in women. It increased the percentage (Azziz 2003; Martin et al. 2008; Alsantali and
of hair follicles in anagen and may cause some Shapiro 2009; Lapidoth et al. 2010). These range
regrowth, but is probably most effective in preventing from simple cosmetic treatments to oral pharma-
further progression (Dawber and Van Neste 1995; ceutical medication.
Vexiau et al. 2002). Since cyproterone acetate is
unavailable in the USA, spironolactone and high- 7.6.2.1 Non-hormonal approaches
dose cimetidine have been used as alternative Cosmetic treatments such as bleaching and depilatory
antiandrogens. measures, such as shaving, waxing or depilatory
Finasteride, a 5a-reductase type 2 inhibitor, was creams, are common, but only remove hair or reduce
developed to treat androgen-potentiated prostate dis- the amount or appearance of hair temporarily. Elec-
orders and is now available as an oral treatment for trolysis, with the aim of permanent removal by killing
androgenetic alopecia in men in many countries at a the dermal papilla and germinative epithelium/stem
lower dose of 1 mg/d. Clinical trials demonstrated cells is the most established long-lasting treatment,
significant effects on stimulating hair regrowth in but it is expensive, time consuming and may cause
younger men with mild to moderate hair loss (Kauf- scarring. Photoepilation, using laser or intense pulse
man et al. 1998). More recently effects have also been light (IPL) treatment, is a more recently introduced
seen in older men (Kaufman et al. 2008a; 2008b). alternative to electrolysis (Sanchez et al. 2002; Alsan-
Even if hair did not regrow, balding progression was tali and Shapiro 2009). A Cochrane review of photo-
frequently halted and no significant adverse effects epilation found that alexandrite and diode lasers were
have been reported. Unfortunately, no effects of finas- more useful, while there was little evidence of effect-
teride have been seen in postmenopausal women with iveness with IPL, longer wavelength (Nd:YAG) or
androgenetic alopecia (Price et al. 2000); use in pre- ruby lasers (Haedersdal and Gotzsche 2006).
menopausal women requires ensuring against contra- A hair growth inhibitor, eflornithine
ception as for antiandrogens. Topical routes of hydrochloride, is available by prescription as a topical
application of finasteride in liposomes have also been cream as Vaniqa, or under other names around the
studied but are not yet available clinically (Kumar world. It is an irreversible inhibitor of the enzyme
et al. 2007). Interestingly, response to finasteride in ornithine decarboxylase which is approved by the
women has recently been shown to depend on genetic FDA (USA) for the reduction of unwanted facial hair
variations in the androgen receptor gene, bringing in women. It is used alone or as an adjuvant to laser
in a whole new area of epigenetics to alopecia therapy (Lapidoth et al. 2010).
treatment (Keene and Goren 2011). Previously, Tang
et al. (2003) reported that response to finasteride in 7.6.2.2 Endocrine-based treatments
men was correlated to the ability of their follicle Oral contraceptives are generally considered the first-
dermal papilla cells to produce IGF-1. Both these line therapy for hirsutism in premenopausal women
results show how important it is to understand the (Martin et al. 2008), in the form of an estrogen,
mechanism of androgen action in hair follicles to frequently ethinyl estradiol, in combination with
develop new treatments. A short trial of dutasteride, a progestin. The progestin used should not have
a dual inhibitor of both 5a-reductase types 1 and 2 androgenic properties, and a combination with an

168
Chapter 7: Androgens and hair

antiandrogenic form, which should compete for the contact with the blood or is specific for hair follicles
androgen receptor in the follicle, such as cyproterone is not yet available. Further research on the biology of
acetate or drospirenone, is recommended (Alsantali androgen action in the hair follicle may facilitate its
and Shapiro 2009). Oral contraceptives act by sup- development.
pressing luteinizing hormone secretion and thereby
inhibiting ovarian androgen biosynthesis, increasing 7.7 Key messages
SHBG production (which decreases serum free  Androgens are the main regulator of human hair
androgens), and causing some reduction in androgen growth and responsible for initiating clinical
synthesis by the adrenals. conditions including androgenetic alopecia and
If the oral contraceptives are insufficient, anti- hirsutism.
androgens are recommended (Martin et al. 2008).  Androgens have paradoxically different effects
on hair follicles depending on their body site.
The most common antiandrogen treatment, outside
They can stimulate the formation of large hairs,
the USA, is cyproterone acetate, given with estrogen e.g. beard, axilla; have no effect, e.g. eyelashes; or
if the woman is premenopausal. It is used at higher inhibit follicles on the scalp.
doses, but must still be balanced with estrogens if  All effects are gradual.
pregnancy is a possibility. Spironolactone, an aldos-  Common androgen-potentiated disorders of hair
terone antagonist, known to have antiandrogen growth, including hirsutism in women and
activity, or flutamide, a pure non-steroidal antian- androgenetic alopecia in both sexes, particularly in
drogen, can be used as alternatives (Swiglo et al. men, cause widespread psychological distress due
2008); flutamide is not recommended as a front-line to the importance of hair in human
therapy due to hepatotoxicity (Martin et al. 2008; communication.
Alsantali and Shapiro 2009). Patients have to be  Androgen receptors are necessary for all
androgen-dependent growth and 5a-reductase
well motivated because hair growth on the face
type 2 for most hair, except for female patterns of
generally takes at least nine months before a notice- axillary and pubic hair, even in men.
able effect occurs, although any acne will be cleared  The action of androgens on human hair follicles
in a couple of months and effects on thigh hair demonstrates several paradoxes: contrasting
growth will be seen in four to six months (Sawers effects in different sites; major differences in the
et al. 1982). Facial responses are seen first on the persistence of stimulatory effects depending on
sides of the face and last on the upper lip, in reverse body region; a varying requirement for the
order to the appearance of facial hair in men formation of 5a-dihydrotestosterone even
(personal observations). amongst follicles exhibiting increased growth.
Finasteride (see Section 7.6.2.1) has also been used Since these are all site-related and retained on
transplantation, these indicate intrinsic
for hirsutism with some success (Lakryc et al. 2003;
differences within follicles, presumably
Alsantali and Shapiro 2009). This seems logical, as
determined during embryonic development.
5a-reductase type 2 is necessary for male pattern body  The current model for androgen action in the hair
hair growth (see Section 7.4.1). Contraception is still follicle proposes that androgens act via the cells of
required, as with all endocrine treatments, due to the the dermal papilla, altering their production of
potential to affect the development of a male fetus. regulatory paracrine factors such as growth
Metformin, insulin-sensitizing therapy, aimed factors, which then influence the activity of other
at altering the insulin resistance and hence the follicular components, e.g. keratinocytes,
hyperandrogenism often associated with polycystic melanocytes and endothelial cells. The dermal
ovarian disease, has been used clinically (Harborne sheath may also play a role as a direct androgen
et al. 2003). However, insulin-sensitizing drugs target.
including the thiazolidinediones (e.g. rosiglitazone  Treatment for androgen-dependent hair
disorders involves cosmetic, surgical, endocrine
and pioglitazone) do not appear effective (Cosma
and non-endocrine related therapies.
et al. 2008).  A 5a-reductase type 2 inhibitor, finasteride,
Overall, there have been major changes in the and antiandrogens, generally cyproterone
treatment of androgen-potentiated disorders over acetate, are being used to control androgenetic
the last 10 years. The ideal therapy of a uniformly alopecia and hirsutism. Such treatments of
effective, topical treatment which is inactivated on

169
Chapter 7: Androgens and hair

premenopausal women require  Further understanding of the mechanism of


combination with estrogens to ensure androgens in the hair follicle is necessary to
contraception to avoid possible feminization of a enable the development of better treatments,
male fetus. preferably working topically and specific to the
 Treatments may need several months to hair follicle.
show their effects and will need to be used
continually as long as the source of androgens is
present.
 Non-endocrine treatments for hair loss include
Acknowledgements
minoxidil; prostaglandin-related glaucoma drugs I should like to acknowledge the assistance in prepar-
are under investigation. ation of this manuscript by Lucy Scott, and Chris
Bowers and Jenny Braithwaite for the figures.

7.8 References Bergfeld WF (1955) Androgenetic


alopecia: an autosomal dominant
Botchkareva NV, Ahluwalia G,
Shander D (2006) Apoptosis in the
Ahmad W, Faiyaz ul Haque M, disorder. Am J Med 98:955985 hair follicle. J Invest Dermatol
Brancolini V, Tsou HC, ul Haque S, 126:258264
Lam H, Aita VM, Owen J, Birch MP, Lalla SC, Messenger AG
deBlaquiere M, Frank J, Cserhalmi- (2002) Female pattern hair loss. Clin Botchkareva VA, Kishimoto J (2003)
Friedman PB, Leask A, McGrath JA, Exp Dermatol 27:383388 Molecular control of epithelial-
Peacocke M, Ahmad M, Ott J, Blume-Peytavi U, Atkin S, Shapiro J, mesenchymal interactions during
Christiano AM (1998) Alopecia Lavery S, Grimalt R, Hoffmann R, hair follicle cycling. J Invest
universalis associated with a Gieler U, Messenger A (2009) Dermatol Symp Proc 8:4655
mutation in the human hairless European Consensus on the Bradfield RB (1971) Protein
gene. Science 279:720724 evaluation of women presenting deprivation: comparative response
Alsantali A, Shapiro J (2009) with excessive hair growth. Eur of hair roots, serum protein and
Management of hirsutism. Skin J Dermatol 19:597602 urinary nitrogen. Amer J Clin Nutrit
Therapy Lett 14:13 Blume-Peytavi U, Blumeyer A, Tosti A, 24:405410
Finner A, Marmol V, Trakatelli M, Braun-Falco O, Christophers E (1968)
Ando Y, Yamaguchi Y, Hamada K,
Reygagne P, Messenger A; Hair root patterns in male pattern
Yoshikawa K, Itami S (1999)
European Consensus Group alopecia. In: Baccareda-Boy A,
Expression of mRNA for androgen
(2011a) S1 guideline for diagnostic Moretti G, Fray JR (eds)
receptor, 5a-reductase and 17b-
evaluation in androgenetic alopecia Biopathology of Pattern Alopecia.
hydroxysteroid dehydrogenase in
in men, women and adolescents. Br Karger, Basel, pp 141145
human dermal papilla cells. Br
J Dermatol 164:515
J Dermatol 141:840845 Carey AH, Chan KL, Short F, White D,
Blume-Peytavi U, Hillmann K, Dietz E, Williamson R, Franks S (1993)
Aoi N, Inoue K, Kato H, Suga H,
Canfield D, Garcia Bartels N Evidence for a single gene effect
Higashino T, Eto H, Doi K, Araki J,
(2011b) A randomized, single-blind causing polycystic ovaries and male
Iida T, Katsuta T, Yoshimura KJ
trial of 5% minoxidil foam once pattern baldness. Clin Endocrinol
(2012) Clinically applicable
daily versus 2% minoxidil 38:653658
transplantation procedure of dermal
solution twice daily in the treatment
papilla cells for hair follicle Cash TF (1993) Psychological effects of
of androgenetic alopecia in
regeneration. J Tissue Eng Regen Med androgenetic alopecia on women:
women. J Am Acad Dermatol
6:8595. DOI 10.1002/term.400 comparisons with balding men and
65:11261134.e2
Asada Y, Sonoda T, Ojiro M, Kurata S, with female control subjects. J Am
Blume-Peytavi U, Lnnfors S, Acad Dermatol 29:568575
Sato T, Ezaki T, Takayasu S (2001) Hillmann K, Garcia Bartels N
5a-reductase type 2 is constitutively (2011c) A randomized double-blind Cash TF (2009) Attitudes, behaviors, and
expressed in the dermal papilla and placebo-controlled pilot study to expectations of men seeking medical
connective tissue sheath of the hair assess the efficacy of a 24-week treatment for male pattern hair loss:
follicle in vivo but not during topical treatment by latanoprost results of a multinational survey.
culture in vitro. J Clin Endocr Metab 0.1% on hair growth and Curr Med Res Opin 25:18111820
86:28752880 pigmentation in healthy volunteers Chieffi M (1949) Effect of testosterone
Azziz R (2003) The evaluation and with androgenetic alopecia. J Am administration on the beard
management of hirsutism. Obstet Acad Dermatol Aug 27 [Epub ahead growth of elderly males. J Geront
Gynaecol 101:9951007 of print] 4:200204

170
Chapter 7: Androgens and hair

Choudhry R, Hodgins MB, Van Der Ebling FJG (1985) Age changes in to thinning hair. J Soc Psychol
Kwast TH, Brinkman AO, Boersma cutaneous appendages. J Appl 130:209218
WJA (1992) Localisation of Cosmetol 3:243250 Garza LA, Yang CC, Zhao T, Blatt HB,
androgen receptors in human skin Ebling FJG, Johnson E (1959) Hair Lee M, He H, Stanton DC, Carrasco
by immunohistochemistry: growth and its relation to L, Spiegel JH, Tobias JW, Cotsarelis
implications for the hormonal vascular supply in rotated skin G (2011) Bald scalp in men with
regulation of hair growth, sebaceous grafts and transposed flaps in the androgenetic alopecia retains hair
glands and sweat glands. albino rat. J Embry Exp Morph follicle stem cells but lacks CD200-
J Endocrinol 133:467475 9:285293 rich and CD34-positive hair follicle
Cobb JE, White SJ, Harrap SB, Ellis JA Ebling FJG, Hale PA, Randall VA progenitor cells. J Clin Invest
(2009) Androgen receptor copy (1991) Hormones and hair growth. 121:613622
number variation and androgenetic In: Goldsmith LA (ed) Biochemistry Giles GG, Severi G, Sinclair R, English
alopecia: a case-control study. PLoS and Physiology of the Skin, 2nd edn. DR, McCredie MR, Johnson W,
One 4:e5081 Clarenden Press, Oxford, Boyle P, Hopper JL (2002)
Cosma M, Swiglo BA, Flynn DN, Kurtz pp 660696 Androgenetic alopecia and prostate
DM, Labella ML, Mullan RJ, Elamin Elliott K, Stephenson TJ, Messenger cancer: findings from an Australian
MB, Erwin PJ, Montori VM (2008) AG (1999) Differences in hair case-control study. Can Epidemiol
Clinical review: insulin sensitizers follicle dermal papilla volume are 11:549553
for the treatment of hirsutism: a due to extracellular matrix volume Girman CJ, Rhodes T, Lilly FRW Guo
systematic review and metaanalyses and cell number: implications for SS, Siervogel RM, Patrick DL,
of randomized controlled trials. the control of hair follicle size and Chumlea WC (1998) Effects of self-
J Clin Endocr Metab 93:11351142 androgen responses. J Invest perceived hair loss in a community
Cotsarelis C, Sun T, Lavker R (1990) Dermatol 113:873877 sample of men. Dermatology
Label-retaining cells reside in the Ellis JA, Harrap SB (2001) The genetics 197:223229
bulge area of the pilosebaceous unit; of androgenetic alopecia. Clin Gokrem S, Baser NT, Aslan G (2008)
implications for follicular stem cells, Dermatol 19:149154 Follicular unit extraction in hair
hair cycle and skin carcinogenesis. transplantation: personal
Ellis JA, Stebbing M, Harrap SB (1998)
Cell 61:13291337 experience. Ann Plast Surg
Genetic analysis of male pattern
Courtois M, Loussouarn G, Hourseau baldness and the 5a-reductase 60 (2):127133
C, Grollier JF (1995) Ageing and genes. J Invest Dermatol Goodhart DB (1960) The evolutionary
hair cycles. Br J Dermatol 132:8693 110:849853 significance of human hair patterns
Courtois M, Loussouarn G, Hourseau Ellis JA, Stebbing M, Harrap SB and skin coloring. Adv Sci 17:5359
S, Grollier JF (1996) Periodicity in (2001a) Male pattern baldness is not Griffin JE, Wilson JD (1989) The
the growth and shedding of hair. Br associated with established resistance syndromes: 5a-reductase
J Dermatol 134:4754 cardiovascular risk factors in the deficiency, testicular feminisation
Cousen P, Messenger A (2010) Female general population. Clin Sci and related disorders. In: Scriver
pattern hair loss in complete 100:401404 CR, Baudet AL, Sly WS, Valle D
androgen insensitivity syndrome. Br Ellis JA, Stebbing M, Harrap SB (eds) The Metabolic Basis of
J Dermatol 162:11351137 (2001b) Polymorphism of androgen Inherited Disease. McGraw-Hill,
Curran, MP (2009) Bimatoprost: a receptor gene is associated with New York, pp 19191944
review of its use in open angle male pattern baldness. J Invest Ha SJ, Kim JS, Myung JW, Lee HJ, Kim
glaucoma and ocular hypertension. Dermatol 116:452455 JW (2003) Analysis of genetic
Drugs Aging 26:10491071 Ferriman D, Gallwey JD (1961) Clinical polymorphisms of steroid 5a-
Dawber R, Van Neste D (1995) Hair assessment of body hair growth in reductase type 1 and 2 genes in
and Scalp Disorders. Martin Dunitz, women. J Clin Endocr Metab Korean men with androgenetic
London 21:14401447 alopecia. J Dermatol Sci 31:135141
Demark-Wahnefried W, Lesko SM, Flux JEC (1970) Colour change of Haedersdal M, Gotzsche PC (2006)
Conaway MR, Robertson CN, Clark mountain hares (Lepus timidus Laser and photoepilation for
RV, Lobaugh B, Mathias BJ, Strigo scoticus) in north-east Scotland. unwanted hair growth. Cochrane
TS, Paulsa DF (1997) Serum Zool 162:345358 Database Syst Rev 4:CD004684
androgens: associations with Franzoi SL, Anderson J, Frommelt S Hamada K, Randall VA (2006)
prostate cancer risk and hair (1990) Individual differences in Inhibitory autocrine factors
pattening. J Androl 18:495450 mens perceptions of and reactions produced by the mesenchyme-

171
Chapter 7: Androgens and hair

derived hair follicle dermal Randall VA (eds) Hair Research for Hillmer AM, Freudenberg J, Myles S,
papilla may be a key to male the Next Millennium. Elsevier Herms S, Tang K, Hughes DA,
pattern baldness. Br J Dermatol Science, Amsterdam, Brockschmidt FF, Ruan Y,
154:609618 pp 303306 Stonkeking M, Nthen MM (2009)
Hibberts NA, Sato K, Messenger AG, Recent positive selection of a human
Hamada K, Thornton MJ, Liang I,
Randall VA (1996a) Dermal papilla androgen receptor/ectodysplasin A2
Messenger AG, Randall VA (1996)
cells from human hair follicles receptor haplotype and its
Pubic and axillary dermal papilla
secrete factors (eg VEGF) mitogenic relationship to male pattern
cells do not produce 5a-
for endothelial cells. J Invest baldness. Hum Genet 126:255264
dihydrotestosterone in culture.
J Invest Dermatol 106:10171022 Dermatol 106:341 Hoffmann R, Happle R (2000) Current
understanding of androgenetic
Hamilton JB (1942) Male hormone Hibberts NA, Messenger AG, Randall
alopecia. Part II: clinical aspects
stimulation is a prerequisite and an VA (1996b) Dermal papilla cells
and treatment. Eur J Dermatol
incitant in common baldness. Amer derived from beard hair follicles
10:410417
J Anat 71:451480 secrete more stem cell factor (SCF)
in culture than scalp cells or dermal Hsu YC, Pasolli HA, Fuchs E (2011)
Hamilton JB (1946) A secondary sexual Dynamics between stem cells, niche,
fibroblasts. Biochem Biophys Res
character that develops in men but
Commun 222:401405 and progeny in the hair follicle. Cell
not in women upon aging of an 144:92105
organ present in both sexes. Anat Hibberts NA, Howell AE, Randall VA
Rec 94:466467 (1998) Dermal papilla cells Imperato-McGinley J, Gautier T, Cai L,
from human balding scalp hair Yee B, Epstein J, Pochi P (1993) The
Hamilton JB (1951a) Patterned loss of androgen control of sebum
follicles contain higher levels of
hair in man; types and incidence. production. Studies of subjects with
androgen receptors than those from
Ann N Y Acad Sci 53:708728 dihydrotestosterone deficiency and
non-balding scalp. J Endocrinol
Hamilton JB (1951b) Quantitative 156:5965 complete androgen insensitivity.
measurement of a secondary sex J Clin Endocr Metab 76:524528
Hibino T, Nishiyama T (2004) Role of
character and axillary hair. Ann N Y Inui S, Fukuzato Y, Nakasima T,
TGF-b2 in the human hair cycle.
Acad Sci 53:585599 Yoshikawa (2002) Androgen-
J Dermatol Sci 35:918
Hamilton JB (1958) Age, sex and inducible TGF-beta1 from balding
Higgins CA, Richardson, GD, Westagte dermal papilla cells inhibits
genetic factors in the regulation of
GE, Jahoda CA (2009) Exogen epithelial cell growth: a clue to
hair growth in man: a comparison
involves gradual release of the hair understand paradoxical effects of
of Caucasian & Japanese
club fiber in the vibrissa follicle androgen on human hair growth.
populations. In: Montagna W,
model. Exp Dermatol 18:793795 FASEB J 16 (14):19671969
Ellis RA (eds) The Biology of Hair
Growth. Academic Press, New York, Hillmer AM, Kruse R, Macciardi F, Itami S, Kurata S, Takayasu S (1990)
pp 399433 Heyn U, Betz RC, Ruzicka T, 5a-reductase activity in cultured
Propping P, Nothen MM, Cichon S human dermal papilla cells from
Harborne L, Fleming R, Lyall H,
(2002) The hairless gene in beard compared with reticular
Norman J, Sattar N (2003)
androgenetic alopecia: results of a dermal fibroblasts. J Invest Dermatol
Descriptive review of the evidence
systematic mutation screening and a 94:150152
for the use of metformin in
family-based association approach.
polycystic ovary syndrome. Lancet Itami S, Kurata S, Sonada T, Takayasu
Br J Dermatol 146:601608
361:18941901 S (1995a) Interactions between
Hillmer AM, Brockschmidt FF, dermal papilla cells and
Hayes VM, Saveri G, Padilla E, Morris
Hanneken S, Eigelshoven S, Steffens follicular epithelial cells in vitro:
H, Tilley W, Southey M, English D,
M, Flaquer A, Herms S, Becker T, effect of androgen. Br J Dermatol
Sutherland R, Hopper J, Boyle P,
Kortm AK, Nyholt DR, Zhao ZZ, 132:527532
Giles G (2006) 5a-reductase type 2
Montgomery GW, Martin NG,
gene variant associations with Itami S, Kurata S, Takayasu S (1995b)
Mlenisen TW, Alblas MA, Moebus
prostate cancer risk, circulating Androgen induction of follicular
S, Jckel KH, Brcker-Preuss M,
hormone levels and androgenetic epithelial cell growth is mediated via
Erbel R, Reinartz R, Betz RC,
alopecia. Int J Cancer 120:776780 insulin-like growth factor I from
Cichon S, Propping P, Baur MP,
Hibberts NA, Randall VA (1996) Wienker TF, Kruse R, Nthen MM dermal papilla cells. Biochem
Testosterone inhibits the capacity of (2008) Susceptibility variants for Biophys Res Commun 212:988994
cultured balding scalp dermal male pattern baldness on Jackson D, Church RE, Ebling FJG
papilla cells to produce keratinocyte chromosome 20p11. Nat Genet (1972) Hair diameter in female
mitogenic factors. In: Van Neste D, 40:12791281 baldness. Br J Dermatol 87:361367

172
Chapter 7: Androgens and hair

Jahoda CA, Horne KA, Oliver RF Kim WS, Lee HI, Lee JW, Lim YY, Lee practice options for hair removal in
(1984) Induction of hair growth by SJ, Kim BJ, Kim MN, Song KY, Park patients with unwanted facial hair
implantation of cultured dermal WS (2011) Fractional using combination therapy with
papilla cells. Nature 311 photothermolysis laser treatment of laser: guidelines drawn up by an
(5986):560562 male pattern hair loss. Dermatol expert working group. Dermatology
Jansen VA, Van Baalen M (2006) Surg 37:4151 221:3442
Altruism through beard Kitagawa T, Matsuda KI, Inui S, Leavitt M, Charles G, Heyman E,
chromodynamics. Nature Takenaka H, Katoh N, Itami S, Michaels D (2009) HairMax
440:663666 Kishimoto S, Kawata M (2009) LaserComb laser phototherapy
Jave-Suarez LF, Langbein L, Winter H, Keratinocyte growth inhibition device in the treatment of male
Praetzel S, Rogers MA, Schweizer J through the modification of Wnt androgenetic alopecia: a
(2004) Androgen regulation of the signaling by androgen in balding randomized, double-blind,
human hair follicle: the type 1 hair dermal papilla cells. J Clin sham device-controlled, multicentre
keratin hHa7 is a direct target gene Endocrinol Metab 94:12881294 trial. Clin Drug Investig
in trichocytes. J Invest Dermatol 29:283292
Kligman AM (1959) The human hair
122:555564 cycle. J Invest Dermatol 33:307316 Lesko SM, Rosenberg L, Shapiro S
Jenkins JS, Ash S (1973) The (1993) A case-control study of
Kligman AM (1988) The comparative baldness in relation to myocardial
metabolism of testosterone by histopathology of male-pattern
human skin in disorders of hair infarction in men. JAMA
baldness and senescent baldness. 269:9981003
growth. J Endocrinol 59:345351 Clin Dermatol 6:108118
Kaufman KD, Olsen EA, Whiting D, Ludwig E (1977) Classification of the
Konig A, Happle R, Tchitcherina E, types of androgenetic alopecia
Savi R, De Villez R, Bergfeld W; Schaefer JR, Sokolowski P, Kohler
Finasteride Male Pattern Hair Loss (common baldness) occurring in
W, Hoffmann R (2000) An X-linked the female sex. Br J Dermatol
Study Group (1998) Finasteride in gene involved in androgenetic
the treatment of men with 97:247254
alopecia: a lesson to be learned from
androgenetic alopecia. J Am Acad adrenoleukodystrophy. Lui F, Uchugonova A, Kimura H,
Dermatol 39:578589 Dermatology 200:213218 Zhang C, Zhao M, Zhang L, Koenig
Kaufman KD, Rotonda J, Shah AK, K, Duong J, Aki R, Saito N, Mii S,
Kumar R, Singh B, Bakshi G, Katare Amoh Y, Katsuoka K, Hoffman RM
Meehan AG (2008a) Long-term OP (2007) Development of
treatment with finasteride 1 mg (2011) The bulge area is the major
liposomal systems of finasteride for hair follicle source of nestin-
decreases the likelihood of topical applications: design,
developing further visible hair loss expressing pluripotent stem cells
characterization, and in vitro which can repair the spinal cord
in men with androgenetic alopecia evaluation. Pharm Dev Technol
(male pattern hair loss). Eur compared to the dermal papilla. Cell
12:591601 Cycle 10:830839
J Dermatol 18:400406
Kwack, MH, Sung YK, Chung EJ Lynfield YL (1960) Effect of pregnancy
Kaufman KD, Girman CJ, Round EM,
(2008) Dihydrotestosterone- on the human hair cycle. J Invest
Johnson-Levonas AO, Shah AK,
inducible dickkopf 1 from balding Dermatol 35:323327
Rotonda J (2008b) Progression of
dermal papilla cells causes apoptosis
hair loss in men with androgenetic Marshall WA, Tanner JM (1969)
in follicular keratinocytes. J Invest
alopecia (male pattern hair loss): Variations in pattern of pubertal
Dermat 128:262269
long-term (5-year) controlled change in girls. Arch Dis Child
observational data in placebo- Lakryc EM, Motta EL, Soares JM Jr, 44:291303
treated patients. Eur J Dermatol Haider MA, de Lima GR, Baracat
Marshall WA, Tanner JM (1970)
18:407411 EC (2003) The benefits of
Variations in the pattern of pubertal
finasteride for hirsute women with
Keene S, Goren (2011) A therapeutic changes in boys. Arch Dis Child
polycystic ovary syndrome or
hotline. Genetic variations in the 45:1323
idiopathic hirsutism. Gynaecol
androgen receptor gene and
Endocrinol 17:5763 Martin KA, Chang RJ, Ehrmann DA,
finasteride response in women Ibanez L, Lobo RA, Rosenfield RL,
with androgenetic alopecia Lambert G (1961) The Conquest of Shapiro J, Montori VM, Swiglo BA
mediated by epigenetics. Dermatol Baldness. The Wonderful Story of (2008) Evaluation and treatment of
Ther 24:296300 Hair. Souvenir Press, London hirsutism in premenopausal
Keogh EV, Walsh RJ (1965) Rate of Lapidoth M, Dierickx C, Lanigan S, women: an Endocrine Society
greying of human hair. Nature Paasch U, Campo-Voegeli A, Dahan clinical practice guideline. J Clin
207:877878 S, Marini L, Adatto M (2010) Best Endocrinol Metab 93:11051120

173
Chapter 7: Androgens and hair

Messenger AG, Rundegren J (2004) W, Dobson RL (eds) Hair Technology, Skinterface, Brussels,
Minoxidil: mechanisms of action Growth, Advances in Biology of pp 7592
on hair growth. Br J Dermatol Skin, Vol. 9. Pergamon Press, Randall VA (2005) Physiology and
150:186194 Oxford, pp 99108 pathophysiology of androgenetic
Midorikawa T, Chikazawa T, Orentreich N, Durr NP (1982) Biology alopecia. In:Degroot LJ, Jameson JL
Yoshino T, Takada K, Arase S of scalp hair growth. Clin Plas Surg (eds)Endocrinology, 5th edn.
(2004) Different gene expression 9:197205 W B Saunders Co, Philadelphia,
profile observed in dermal papilla Section XIV, Burger HG (ed) Male
Osbourn D (1916) Inheritance of
cells related to androgenic alopecia Reproduction, Chapter 178, pp
baldness. J Hered 7:347355
by DNA macroarray analysis. 32953309
J Dermatol Sci 36:2532 Philpott MP (2000) The roles of growth
Randall VA (2007) Hormonal
factors in hair follicles:
Miranda BH, Tobin DJ, Sharpe DT, regulation of hair follicles exhibits a
Randall VA (2010) Intermediate investigations using cultured hair
biological paradox. Semin Cell Dev
follicles. In: Camacho F, Randall
hair follicles: a new more clinically Biol 18:274285
relevant model for hair growth VA, Price VH (eds) Hair and its
Disorders: Biology, Research and Randall VA (2008) Androgens and hair
investigations. Br J Dermatol
Management. Martin Dunitz, growth. Dermatol Ther 21:314328
163:287295
London, pp 103113 Randall VA (2010) Molecular basis of
Nishimura EK (2011) Melanocyte stem
Price VH, Roberts JL, Hordinsky M, androgenetic alopecia In: Treb
cells: a melanocyte reservoir in hair
Olsen EA, Savin R, Bergfeld W, RM, Tobin DJ (eds) Aging Hair.
follicles for hair and skin
Fiedler V, Lucky A, Whiting DA, Springer-Verlag, Berlin, Chapter 2,
pigmentation. Pigment Cell
Pappas F, Culbertson J, Kotey P, pp 924
Melanoma Res 24:401410
Meehan A, Waldstreicher J (2000) Randall VA, Botchkareva NV (2009)
Norwood OT (1975) Male pattern Lack of efficacy of finasteride in The biology of hair growth. In:
baldness, classification and postmenopausal women with Ahluwalia GA (ed) Cosmetic
incidence. South Med J androgenetic alopecia. J Am Acad Applications of Laser and Light
68:13591365 Dermatol 43:768776 Based Systems. William Andrew,
Obana N, Chang C, Uno H (1997) Rabbani P, Takeo M, Chou W, Myung Norwich, NY, pp 335
Inhibition of hair growth by P, Bosenberg M, Chin L, Taketo Randall VA, Ebling FJG (1991)
testosterone in the presence of MM, Ito M (2011) Coordinated Seasonal changes in human hair
dermal papilla cells from the frontal activation of Wnt in epithelial and growth. Br J Dermatol 124:146151
bald scalp of the post-pubertal melanocyte stem cells initiates
stump-tailed macaque. pigmented hair regeneration. Cell Randall VA, Thornton MJ, Hamada K,
Endocrinology 138:356361 10 (145):941955 Redfern CPF, Nutbrown M, Ebling
Olsen EA, Dunlap FE, Funicella T, FJG, Messenger AG (1991)
Randall VA (1994) Androgens and Androgens and the hair follicle:
Koperski JA, Swinehart JM, Tschen
human hair growth. Clin Endocrinol cultured human dermal papilla cells
EH, Trancik RJ (2002)
40:439457 as a model system. Ann N Y Acad
A randomized clinical trial of 5%
topical minoxidil versus 2% topical Randall VA (2000a) Androgens: the Sci 642:355375
minoxidil and placebo in the main regulator of human hair Randall VA, Thornton MJ, Messenger
treatment of androgenetic alopecia growth. In: Camacho F, Randall AG (1992) Cultured dermal papilla
in men. J Am Acad Dermatol VA, Price VH (eds) Hair and its cells from androgen-dependent
47:377385 Disorders: Biology, Research and human follicles (e.g. beard) contain
Management. Martin Dunitz, more androgen receptors than those
Olsen EA, Hordinsky M, Whiting D,
London, pp 6982 from non-balding areas.
Stough D, Hobbs S, Ellis ML,
Wilson T, Rittmaster RS (2006) The Randall VA (2000b) The biology of J Endocrinol 133:141147
importance of dual 5alpha- androgenetic alopecia. In: Camacho Randall VA, Jenner T, Hibberts N, De
reductase inhibition in the F, Randall VA, Price VH (eds) Hair Oliveira I, Vafaee T (2008) Stem cell
treatment of male pattern hair loss: and its Disorders: Biology, Research factor/c-kit signalling in normal and
results of a randomized placebo- and Management. Martin Dunitz, androgenetic alopecia hair follicles.
controlled study of dutasteride London, pp 123136 J Endocrinol 197:114
versus finasteride. J Am Acad Randall VA (2003) Biological effects of Rendl M, Lewis L, Fuchs E (2005)
Dermatol 55:10141023 androgens on the hair follicle: Molecular dissection of
Orentreich N (1969) Scalp hair experimental approaches. In: Van mesenchymal-epithelial interactions
replacement in man. In: Montagna Neste D (ed) Hair Science and in the hair follicle. PLoS Biol 3:331

174
Chapter 7: Androgens and hair

Reynolds AJ, Jahoda CAB (2004) topics in Endocrinology. John eyelash characterization. Br
Cultured human and rat tooth Wiley & Sons, Ltd., Chichester, J Dermatol 162:304310
papilla cells induce hair follicle pp 145168 Thomson AA, Foster BA, Cunha GR
regeneration and fiber growth. Setty LR (1970) Hair patterns of the (1997) Analysis of growth factor
Differentiation 72:566575 scalp of white and Negro males. and receptor mRNA levels during
Reynolds AJ, Lawrence C, Cserhalmi- Amer J Phys Anthrop 33:4955 development of the rat seminal
Friedman PB, Christiano AM, Shapiro J, Kaufman KD (2003) Use of vesicle and prostate. Development
Jahoda CAB (1999) Trans-gender finasteride in the treatment of men 124:24312439
induction of hair follicles. Nature with androgenetic alopecia (male Thornton MJ, Laing I, Hamada K,
402:3334 pattern hair loss). J Invest Dermatol Messenger AG, Randall VA (1993)
Richards JB, Yuan X, Geller F, Symp Proc 8:2023 Differences in testosterone
Waterworth D, Bataille V, Glass D, Shimomura Y, Christiano AM (2010) metabolism by beard and scalp hair
Song K, Waeber G, Vollenweider P, follicle dermal papilla cells. Clin
Biology and genetics of hair.
Aben KK, Kiemeney LA, Walters B, Ann Rev Genomics Hum Genet Endocrinol 39:633639
Soranzo N, Thorsteinsdottir U, 11:109132 Thornton MJ, Hamada K, Messenger
Kong A, Rafnar T, Deloukas P, AG, Randall VA (1998) Beard, but
Sulem P, Stefansson H, Stefansson Shorter K, Farjo NP, Picksley SM,
not scalp, dermal papilla cells
K, Spector TD, Mooser V (2008) Randall VA (2008) Human hair
secrete autocrine growth factors in
Male pattern baldness susceptibility follicles contain two forms of ATP
response to testosterone in vitro.
locus at 20p11. Nat Genet sensitive potassium channels, only
J Invest Dermatol 111:727732
40:12821284 one of which is sensitive to
minoxidil. FASEB 22:17251736 Tsuji Y, Denda S, Soma T, Raferty L,
Rogers NE, Avram MR (2008) Medical Momoi T, Hibino T (2003)
treatments for male and female Sonoda T, Asada Y, Kurata S,
A potential suppressor of TGF-b
pattern hair loss. J Am Acad Takayasu S (1999) The mRNA for
delays catagen progression in hair
Dermatol 59:547566; quiz protease nexin-1 is expressed in
follicles. J Invest Dermatol Symp
56755686 human dermal papilla cells and its
Proc 8:6568
level is affected by androgen. J Invest
Ross EK, Shapiro J (2005) Management Dermatol 113:308313 Van der Donk J, Passchier J, Knegt-
of hair loss. Dermatol Clin Junk C, Wegen-Keijser MH,
23:227243 Stanford JL, Just JJ, Gibbs M, Wicklund
Nieboer C, Stolz E, Verhage F
KG, Neal CL, Blumenstein BA,
Rushton DH, Ramsay ID, Norris MJ, (1991) Psychological characteristics
Ostrander EA (1997) Polymorphic
Gilkes JJH (1991) Natural of women with androgenetic
repeats in the androgen receptor
progression of male pattern alopecia: a controlled study. Br
gene: molecular markers of
baldness in young men. Clin Exp J Dermatol 125:248252
prostate cancer risk. Cancer Res
Dermatol 16:188192 57:11941198 Venning VA, Dawber R (1988)
Rutberg SE, Kolpak ML, Gourley JA, Patterned androgenic alopecia.
Swiglo BA, Cosma M, Flynn DN, Kurtz J Amer Acad Dermatol 18:
Tan G, Henry JP, Shander D (2006) DM, Labella ML, Mullan RJ, Erwin
Differences in expression of 10731077
PJ, Montori VM (2008) Clinical
specific biomarkers distinguish review: antiandrogens for the Vexiau P, Chaspoux C, Boudou P, Fiet
human beard from scalp dermal treatment of hirsutism: a systematic J, Jouanique C, Hardy N, Reygagne
papilla cells. J Invest Dermatol review and metaanalyses of P (2002) Effects of minoxidil 2% vs.
126:25832595 randomized controlled trials. J Clin cyproterone acetate treatment on
Saitoh M, Sakamoto M (1970) Endocr Metab 93:11531160 female androgenetic alopecia: a
Human hair cycle. J Invest Dermatol controlled, 12-month randomized
Tang L, Bernardo O, Bolduc C, Lui H, trial. Br J Dermatol 146:992999
54:6581 Madani S, Shapiro J (2003) The
Sanchez LA, Perez M, Azziz R (2002) expression of insulin-like growth Waters JM, Richardson GD, Jahoda CA
Laser hair reduction in the hirsute factor 1 in follicular dermal papillae (2007) Hair follicle stem cells. Semin
patient: a critical assessment. Hum correlates with therapeutic efficacy Cell Dev Biol 18:245254
Reprod Update 8:169181 of finasteride in androgenetic Wehr TA (1998) Effects of seasonal
Sawers RA, Randall VA, Iqbal MJ alopecia. J Am Acad Dermatol changes in daylength on human
(1982) Studies on the clinical and 49:229233 neuroendocrine function. Horm Res
endocrine aspects of antiandrogens. Thibaut S, De Becker E, Caisey L, Baras 49:118124
In: Jeffcoate SL (ed) Androgens and D, Karatas S, Jammayrac O, Pisella Wehr TA, Duncan WC Jr, Sher L,
Antiandrogen Therapy, Current PJ, Bernard BA (2009) Human Aeschbach D, Schwartz PJ, Turner

175
Chapter 7: Androgens and hair

EH, Postolache TT, Rosenthal NE Winter JSD, Faiman C (1973) Zachmann M, Prader A (1970)
(2001) A circadian signal of Pituitary-gonadal relations in Anabolic and androgenetic effect
change of season in patients female children and adolescents. of testosterone in sexually
with seasonal affective disorder. Paed Res 7:948953 immature boys and its dependency
Arch Gen Psychiatry 58:11151116 Wright JL, Page ST, Lin DW, Stanford on GH. J Clin Endocr Metab
Whiting DA (1993) Diagnostic and JL (2010) Male pattern baldness and 30:8595
predictive value of horizontal prostate cancer risk in a population- Zachmann M, Aynsley-Green A,
sections of scalp biopsy specimens based case-control study. Can Prader A (1976) Interrelations
in male pattern androgenetic Epidemiol 34:131135 of the effects of growth hormone
alopecia. J Am Acad Dermatol Yassa M, Saliou M, De Rycke Y, and testosterone in
28:755763 Hemery C, Henni M, Bachaud JM, hypopituitarism. In: Pecile A,
Winter JSD, Faiman C (1972) Thiounn N, Cosset JM, Giraud P Muller EE (eds) Growth
Pituitary-gonadal relations in male (2011) Male pattern baldness and Hormone and Related Peptides.
children and adolescents. Paed Res the risk of prostate cancer. Ann Excerpta Medica, Amsterdam,
6:125135 Oncol 22:18241827 pp 286290

176
Chapter
Testosterone and bone

8 Dirk Vanderschueren, Mieke Sinnesael, Evelien Gielen,


Frank Claessens, and Steven Boonen

8.1 Introduction 177 8.5 Skeletal effects of testosterone replacement in


8.2 Testosterone deficiency and male elderly men 182
osteoporosis 178 8.6 Skeletal effects of other hormonal agents such
8.2.1 Testosterone deficiency and senile as SERMs, SARMs, non-aromatizable androgens
osteoporosis 178 and aromatase inhibitors in men 182
8.2.2 Testosterone deficiency and idiopathic 8.7 Skeletal effects of other non-hormonal anti-
male osteoporosis 179 osteoporotic drugs in hypogonadal men 183
8.2.3 Testosterone deficiency and secondary 8.8 Preclinical studies of sex steroid replacement in
osteoporosis 180 male rodents 183
8.3 Testosterone deficiency and areal bone 8.9 Potential mechanism of androgen receptor-
density 180 mediated androgen action 184
8.4 Skeletal effects of testosterone replacement in 8.10 Key messages 185
hypogonadal men 181 8.11 References 185

8.1 Introduction considered to protect against osteoporosis and may


Osteoporosis is an important and increasing health therefore potentially explain at least partly the
problem, especially in the elderly: fractures are a con- lower male fracture risk (Khosla et al. 2008). In 1947,
siderable burden for society because of associated eco- Fuller Albright already demonstrated that administra-
nomic costs, morbidity as well as mortality (Center tion of testosterone induced a positive calcium balance
et al. 1999; Ensrud et al. 2000; Empana et al. 2004; in both postmenopausal women and eugonadal men
Fechtenbaum et al. 2005; Lippuner et al. 2005; Maravic (Reifenstein and Albright 1947). Moreover, animal
et al. 2005; Orsini et al. 2005). Osteoporosis is also a studies support this concept of anabolic as well as
gender-related disease: although age-adjusted mortal- antiresorptive skeletal action of androgens: androgens
ity after hip fracture is higher in men than in women, for instance stimulate bone formation during growth
fewer men are affected by osteoporosis (Center et al. and inhibit bone resorption after growth in orchidec-
1999; Kanis et al. 2000; Johnell and Kanis 2006). tomized male rodents (Vanderschueren et al. 2004). In
Possible explanations for this gender difference in hypogonadal men testosterone replacement inhibits
fracture risk are that men have broader and therefore bone resorption (Basaria and Dobs 2001). Therefore,
stronger bones than women and also in contrast to potential beneficial skeletal effects of androgen and/or
postmenopausal women do not experience acceler- SARM replacement in elderly men with low-normal or
ated estrogen deficiency related to bone loss (Seeman borderline low testosterone concentrations have
2003). In this context, a question arises: to what extent received growing attention during the last decades, as
does higher male lifelong androgen secretion contrib- illustrated by a rising number of publications on tes-
ute to these gender-related differences of bone devel- tosterone and bone in Pubmed (19701990: 818 pub-
opment and loss? Androgens are traditionally lications; 19902011: 2421 publications).

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

177
Chapter 8: Testosterone and bone

However, testosterone is also a prohormone of largely explained by estrogen deficiency indeed rep-
estradiol. Many, if not all bone-sparing actions of resents not a single, but a number of disorders. The
testosterone may therefore be explained by conver- impact of changes in serum testosterone on the
sion of androgens into estrogens and subsequent pathophysiology may therefore differ according to
stimulation of the estrogen receptors, which are the etiology of these different forms of male
also in addition to androgen receptors present osteoporosis.
in male bone cells. Indeed, since 1994 several case
reports of men suffering from severe osteoporosis 8.2.1 Testosterone deficiency and senile
despite normal or high testosterone concentrations
have been reported (Lanfranco et al. 2008; Smith osteoporosis
et al. 2008; Zirilli et al. 2008). Low bone mass in Senile osteoporosis is characterized by fragility
these men was explained by estrogen not testoster- fractures which occur in very old men (Boonen
one deficiency, either caused by an inactivating et al. 1997). Recently the relationship between tes-
mutation in the principal ERa (Smith et al. 1994) tosterone and age-related bone loss, bone turnover
and/or in the aromatase enzyme that converts andro- and fractures has been studied very extensively in
gens into estrogens (Morishima et al. 1995; Carani large cohorts of elderly community-dwelling men
et al. 1997; Bilezikian et al. 1998; Bouillon et al. (Barrett-Connor et al. 2000; Goderie-Plomp et al.
2004). Thus, estrogens appear to be involved also in 2004; Amin et al. 2006; Bjornerem et al. 2007;
male, and not only in female, skeletal homeostasis. Meier et al. 2008). Most of these studies found
These observations have fueled the more recent either no or a weak association between age-related
assumption that estrogen and not androgen receptor- changes in serum testosterone and bone loss as
mediated androgen action is necessary for male well as occurrence of fractures (see Table 8.1).
skeletal maintenance. However, according to most of these studies, the
Little is known about the role of endogenous age-related rise of SHBG appears to be related to
testosterone in elderly women. However, older post- bone loss in men. Moreover, serum estradiol pre-
menopausal women with higher circulating concen- dicts bone loss as well as fractures in elderly men.
trations of testosterone also have significantly These observations further strengthened the con-
greater bone mineral density (BMD), independently cept that skeletal action of testosterone is indirectly
of body mass index (BMI), suggesting that circulat- mediated by estrogens. Indeed, in the Osteoporotic
ing testosterone may play a role in the maintenance Fractures in Men Study (MrOS), low estrogen con-
of bone density, even in postmenopausal women centrations appeared to increase fracture risk even
with low serum estradiol concentrations (Rariy in men with high testosterone concentrations
et al. 2011). (Mellstrom et al. 2008). In this study, the relation-
In this chapter, the role of testosterone for male ship between serum estrogen and fracture risk is
bone health will be reviewed. We will also try to not linear. The threshold for estrogen is around a
indicate to what extent testosterone action depends very low concentration of only 16 pg/ml. These
on direct activation of the AR or, alternatively, on low concentrations can only be measured precisely
indirect activation of the ER after aromatization into by mass spectrometry, which probably explains
estrogens. Finally, we will briefly address some of the why the role of estrogens was not established
potential underlying mechanisms of testosterone previously in other studies using less precise
action on bone. immunoassay methods (Goderie-Plomp et al.
2004; Bjornerem et al. 2007).
8.2 Testosterone deficiency Many other risk factors for senile osteoporosis
have currently been identified. A FRAX score,
and male osteoporosis which calculates the absolute risk for major osteo-
The role of testosterone deficiency in the development porotic as well as hip fractures in men, has been
of osteoporosis in men remains controversial. Some developed similarly as in women, since many of the
of the controversy is due to the confusing use of the risk factors for senile osteoporosis are not gender
term osteoporosis in men. Osteoporosis in men in specific (Kanis et al. 2008). In this FRAX score,
contrast to postmenopausal osteoporosis which is estradiol concentrations are not included. Neither

178
Chapter 8: Testosterone and bone

Table 8.1 Overview of cohort studies in men which used fracture as an end-point in relation to serum sex steroid concentrations at start
of the study. The average age at start of the study, the duration of the study, the method of measurement of sex steroid and the main
results are given.

Study Average Number Duration LC-MS/MS Immunoassay Main result


age (years)
Rancho Bernardo 66 352 12 X E: inverse correlation
(Barrett-Connor with fractures
et al. 2000) T: no correlation with
fractures
Dubbo (Meier >60 609 16 X T: no correlation with
et al. 2008) fractures
E: inverse correlation
with fractures
Tromso (Bjornerem 5084 1364 8.4 X T, E, SHBG: no
et al. 2007) correlation with
fractures
Rotterdam (Goderie- 67.6  178 6.5 X T, E, SHBG: no
Plomp et al. 2004) 6.8 correlation with
fractures
Framingham (Amin 71 793 18 X E: inverse correlation
et al. 2006) with fractures
T + E: inverse
correlation with
fractures
MrOS (Mellstrom 75 2639 3.3 X E, SHBG: inverse
et al. 2008) correlation with
fractures
Abbreviations: LC-MS/MS, liquid chromatographytandem mass spectrometry; E, estrogen; T, testosterone; SHBG, sex hormone-binding
globulin.

is frailty, as measured by the frailty index of Rock- 8.2.2 Testosterone deficiency and
wood (Rockwood and Mitnitski 2007) or the oper-
ational definition of Fried (Fried et al. 2001), taken idiopathic male osteoporosis
into account in this fracture risk assessment of Idiopathic male osteoporosis is much less frequent
elderly men. Yet, especially in less-healthy elderly than senile osteoporosis, and is characterized by fra-
men, in whom there is a higher prevalence of hypo- gility fractures, mainly at the spine, occurring in
gonadism, lower testosterone and especially lower middle-aged men (Khosla et al. 1994; Kelepouris
free testosterone are associated with loss of muscle et al. 1995). By definition, the cause of this entity is
strength and may increase the risk for sarcopenia, still unknown (therefore the term idiopathic is
frailty, falls and fractures (Schatzl et al. 2003; used) but it is speculated that low bone mass and
Auyeung et al. 2011; OConnell et al. 2011). In these fragility in these men is caused primarily by low bone
less-healthy men, the relative contribution of age- formation at the level of the individual bone remodel-
related changes in serum testosterone to the risk for ing unit, as evidenced by low wall thickness shown by
osteoporosis and the loss of muscle mass and bone histomorphometry (Pernow et al. 2006; 2009).
strength remains to be further investigated (see also In contrast to postmenopausal osteoporosis, the total
Chapter 16). Indeed, most studies only include rela- number of bone remodeling units on the inner bone
tively healthy community-dwelling men of whom surfaces does not appear to be increased in these men.
only a low percentage is suffering from late-onset Although it cannot be excluded that deficient action
male hypogonadism. and/or secretion of estrogens and IGF-1 may be

179
Chapter 8: Testosterone and bone

involved in the pathophysiology of idiopathic male per volume (or volumetric bone density). Therefore,
osteoporosis, in case control studies serum testoster- broader male bones are denser bones on DXA. Denser
one concentrations are not different in men suffering bones are stronger bones and may explain at least
from idiopathic osteoporosis (Kurland et al. 1997; partly the lower number of fractures in men later
Pernow et al. 2009). in life.
Areal bone density is expressed as T-scores, which
8.2.3 Testosterone deficiency and are standard deviations compared to the mean areal
bone density of a young reference group. Male refer-
secondary osteoporosis ence values are higher than female reference values,
Like postmenopausal women, men with hypogonadal because a young male reference group has a higher
osteoporosis in contrast to men suffering from bone size, whereas volumetric bone density does not
senile or idiopathic osteoporosis do have high bone appear to be different between sexes (Seeman 2001).
turnover (Jackson et al. 1987). High bone turnover is Since higher references of areal bone density reflect
characterized by an increased number of bone remod- lower fracture risk, most authors advocate the more
eling units on the bone surface as well as by a relative conservative use of female instead of male references
imbalance of bone formation versus bone resorption in men, in order not to overestimate fracture risk in
in these units. Therefore, hypogonadal osteoporosis men based on DXA measurements (Kanis and Gluer
clearly is a distinct entity of secondary osteoporosis. 2000; Kanis 2002). Other authors still consider male
Hypogonadal osteoporosis may occur following references appropriate in men, as well as a T-score of
chemical or surgical castration in men suffering from 1 and 2.5 for the respective diagnoses of osteope-
prostatic carcinoma or sexual delinquency (Stepan nia and osteoporosis as in postmenopausal women
et al. 1989; Daniell 1997; Wei et al. 1999). It is well (Kamel 2005; Baim et al. 2008). The use of female
established that chemical and surgical castration versus male references has an impact on the number
induces rapid bone loss as well as fractures. The of men being diagnosed as having either osteopenia
fracture risk in these men is very high and increases or osteoporosis (Holt et al. 2002). In the recent FRAX
early after castration (Melton et al. 2011). Therefore, scores the same absolute density measurements for
hypogonadal osteoporosis clearly illustrates that sex the femoral neck are used for men and women.
steroids are essential for the maintenance of skeletal Low areal bone density in men may result from
integrity in men as well as in women. However, this deficient peak bone mass acquisition. There is a
does not answer the question of to what extent bone strong genetic component related to the interindivi-
loss is related to either testosterone or estrogen defi- dual variation of bone mineral gain during growth, as
ciency, or both. Testosterone deficiency may also shown in twin studies and comparative studies of
occur in other secondary forms of osteoporosis such bone density of father and sons as well as brothers
as steroid-induced osteoporosis, but its relative con- (Pelat et al. 2007). Besides genetic factors, androgens
tribution to the pathophysiology of bone loss in these are also involved in the increase of bone size during
men is less well established (Reid 1998). puberty (see animal data or preclinical studies (Call-
ewaert et al. 2010a; 2010b)). Bone size and therefore
8.3 Testosterone deficiency and strength and area are relatively higher in men com-
pared to women which at least partially may
areal bone density account for skeletal sexual dimorphism. Therefore,
Areal bone density, measured by dual-emission X-ray although age-related changes in testosterone in elderly
absorptiometry (DXA), is used as a proxy for osteo- men may not contribute to fracture risk later in life,
porosis in men as in women. Indeed, low areal bone normal secretion and action of testosterone at
density is associated with fracture risk in both sexes younger age is required for the relative gender-
(De Laet et al. 1997; 1998). Areal bone density is specific protection against osteoporosis in the
greater in men than in women because it reflects not elderly. Nevertheless, most men with decreased bone
only volumetric bone density but also bone geometry. density do not have lower testosterone concentrations
Areal bone density is indeed a two-dimensional pro- in case control studies (Meier et al. 1987; Khosla et al.
jection obtained by DXA of the three-dimensional 1998; Rapado et al. 1999; Araujo et al. 2008). In
bone, which reflects bone size as well as bone density contrast, lower average estradiol not testosterone

180
Chapter 8: Testosterone and bone

concentrations are reported in some (Khosla et al. complete model of hypogonadism since they have
1998; Araujo et al. 2008) but not all of these studies very low residual testosterone concentrations (Finkel-
(Meier et al. 1987). Indeed, not only testosterone but stein et al. 1987). Moreover, in these patients, hypo-
also estradiol is important for the acquisition of areal gonadism occurs during puberty, which is a very
bone density and/or maintenance (Callewaert et al. critical period for skeletal growth and development
2010a). Lower estradiol concentrations in men seem (Finkelstein et al. 1999). Severe sex steroid deficiency
to be associated with wider medullary inner bone area testosterone, estradiol or both may inhibit their
and thinner cortices, suggesting that estradiol may peak bone mass acquisition, thereby explaining the
inhibit net endocortical bone resorption during severe deficit of areal bone density, especially in those
growth (Vermeulen et al. 2002). Estrogen replace- men with open epiphyses at time of diagnosis. Areal
ment in one adult patient suffering from androgen bone density in these adolescent men is also low
insensitivity increased cortical bone area by reducing because it reflects mainly a two-dimensional projec-
the inner endocortical surface, without effect on tra- tion of their small growth-delayed bones. Moreover,
becular bone parameters, as shown by peripheral not only secretion but even timing of testosterone
computerized tomography (pQCT) (Taes et al. secretion during puberty may impact on areal bone
2009). Interestingly, also in one aromatase-deficient density, as shown in studies in patients with delayed
adolescent man the increase of areal bone density puberty (Finkelstein et al. 1992; Bertelloni et al. 1995).
following estrogen replacement was related to an These patients also have lower areal bone density at
increase of cortical not trabecular bone pQCT adult age. Their lower areal bone density may again be
parameters, but this increase was explained by an explained by growth failure, since volumetric in con-
increase of the outer and not of the inner cortical trast to areal bone density was not different from
bone surface (Bouillon et al. 2004). Therefore, these controls at least in one study (Bertelloni et al. 1998).
few case reports suggest that estrogen has more effect The potential impact of early low-dose testosterone
on cortical than on trabecular growing bone in men. replacement on future areal bone density has not been
Nevertheless, the relative contribution of AR-medi- well documented in these patients suffering from
ated versus estrogen action on bone acquisition delayed puberty. Some studies have advocated the
remains not fully established in humans, in contrast use of aromatase inhibitors in combination with
to animal studies (see Section 8.9). low-dose testosterone replacement in order to further
adult height in boys suffering from delayed puberty,
8.4 Skeletal effects of testosterone but this may be at a cost and risk of normal peak bone
mass acquisition due to induction of estrogen defi-
replacement in hypogonadal men ciency (Dunkel and Wickman 2002).
Not only the terminology of osteoporosis, but also of The impact of testosterone deficiency on bone in
that of hypogonadism, is somewhat confusing in the patients with less severe and complete types of hypo-
literature on androgens and bones. Many hypogona- gonadism is not well established. Klinefelter syndrome
dal men included in studies looking at associations represents one of the most frequent forms of hypergo-
between bone density and testosterone replacement nadotropic hypogonadism in men. Patients with
still have significant residual testosterone secretion, Klinefelter syndrome frequently have important
which is in contrast to the more severe estradiol residual testosterone secretion, and may also have
deficiency in postmenopausal women or to the sharp higher estradiol concentrations which may impact on
decline of testosterone observed in men following their skeletal development as well. Moreover, some of
chemical or surgical castration. The threshold of the skeletal deficit reported in Klinefelter patients may
testosterone and/or estradiol concentrations below not be related to hormonal but directly to the under-
which bone resorption is increased is not well lying chromosomal abnormality (Ferlin et al. 2011).
defined in hypogonadal men, but may be below the Nevertheless, testosterone replacement clearly
traditional testosterone threshold of two standard increases and/or maintains areal bone density in
deviations below the mean concentration in young hypogonadal men with well-documented underlying
adult men. testicular or hypothalamic-pituitary dysfunction
Patients with isolated hypogonadotropic hypogo- (Katznelson et al. 1996; Behre et al. 1997). Areal bone
nadism (IHH) represent the most severe and density is therefore considered as a well-accepted and

181
Chapter 8: Testosterone and bone

efficient measurement to evaluate the long-term especially in the elderly (Anderson et al. 1997). In
impact of testosterone replacement in these patients. accordance with this assumption, the effect of testos-
However, testosterone replacement studies are uncon- terone on bone density was only significant for im
trolled because of obvious ethical reasons. Moreover, and not for transdermal replacement in a meta-analy-
these studies include hypogonadal men with mixed sis of studies in elderly men (Fabbri et al. 2007).
causes of hypogonadism as well as more or less severe Moreover, in this meta-analysis, the effect of im
forms of hypogonadism. In addition, long-term bone replacement only significantly increased lumbar and
data on testosterone replacement in hypogonadal not femoral density. The only study that reported an
men are retrospective. Finally, different modes of increase of both lumbar and femoral density
androgen replacement (intramuscular versus trans- following testosterone replacement (with or without
dermal) may have differing impacts on bone. For finasteride) included elderly men with lower testoster-
instance, im testosterone replacement increases both one at baseline, which may explain its greater effect
estradiol and testosterone concentrations, and often (Page et al. 2005). Another study could not show a
above the average physiological range (Anderson significant increase of lumbar and femoral bone dens-
et al. 1997). Therefore, effects of testosterone replace- ity following transdermal testosterone in elderly men
ment on bone may be related to estradiol and/or with low testosterone concentrations at baseline; how-
(higher) testosterone concentrations. ever, a positive trend towards a significant improve-
ment of bone density was found in those men with the
8.5 Skeletal effects of testosterone lowest testosterone levels at inclusion (below 200 ng/dl)
(Snyder et al. 1999). Inclusion of elderly men with
replacement in elderly men concentrations above this threshold may therefore
The potential benefit of testosterone replacement on explain the limited benefit of testosterone on bone
bone in elderly men remains a matter of debate. The density in these studies.
administration of testosterone to hypogonadal men
(especially after chemical and surgical castration)
inhibits bone resorption and maintains bone mass,
8.6 Skeletaleffectsofother hormonal
whereas its effect in elderly men with borderline low agents such as SERMs, SARMs, non-
testosterone or low-normal testosterone concentra- aromatizable androgens and aromatase
tions is more controversial (Isidori et al. 2005).
Recent studies by Kenny et al. (2001; 2002) and inhibitors in men
Crawford et al. (2003) for instance showed no to Given the potential effect of estradiol action on male
minor effects on areal bone density after 12 months bone, selective estrogen receptor modulators (SERMs)
of treatment. One of the problems which may explain may be considered as osteoporosis therapy. However,
some of this controversy is the inclusion of elderly only limited data are available with respect to the
men with only minor or no testosterone deficiency. action of SERMs in men. Raloxifene significantly
Baseline testosterone concentrations of these elderly lowered the decrease in bone density in prostate car-
men are often only borderline and frequently above cinoma patients following chemical castration (Smith
200 ng/dl. Furthermore, studies that have investigated et al. 2004). Raloxifene also had significant effects on
bone end-points mainly focused on areal bone dens- bone turnover markers in elderly men, however with-
ity. None of these studies were powered to investigate out data on bone density (Doran et al. 2001).
fracture risk, which requires much greater numbers of Selective androgen receptor modulators (SARMs),
patients. Indeed, sample size of most of these studies which are the counterpart of SERMs, are another
was small, and duration of follow-up during testoster- potential alternative for testosterone but so far only
one therapy relatively short. In addition, different evaluated in preclinical animal studies (Gao et al.
modes of testosterone replacement were used, which 2005; Kearbey et al. 2009).
makes it difficult to compare studies. As mentioned Administration of the non-aromatizable androgen
before, im testosterone replacement induces higher dihydrotestosterone (DHT) gel to elderly men did not
concentrations of testosterone and estradiol than significantly affect the bone turnover marker osteocalcin,
transdermal testosterone substitution, which may but other more relevant bone end-points such as areal
explain a greater effect of the former administration bone density are not available (Ly et al. 2001).

182
Chapter 8: Testosterone and bone

Administration of the aromatase inhibitor anastro- into estrogen and activation of the ERa. For instance,
zole does increase testosterone levels, but decreases periosteal bone formation is clearly mediated by the
both estradiol concentrations and areal bone mineral AR, as shown by lack of effect of high-dose testoster-
density in elderly men (Burnett-Bowie et al. 2009). one in androgen-resistant mice, in contrast to a stimu-
Therefore, the use of an aromatase inhibitor as alter- latory action in corresponding wild types (Venken
native strategy to increase testosterone levels in elderly et al. 2006). A lower concentration of estrogens stimu-
men may have unwanted side-effects on bone. lates GH-IGF-1 and thereby also periosteal as well as
longitudinal bone formation; whereas higher concen-
trations of estrogens induce growth plate closure in
8.7 Skeletal effects of other non- both sexes (Vanderschueren et al. 2005).
hormonal anti-osteoporotic drugs in The dual mode of action of testosterone on tra-
becular bone is also confirmed during and after
hypogonadal men growth (Callewaert et al. 2009; 2010a). The number
Antiresorptive, anti-osteoporotic drugs such as the of trabeculae is only maintained in the presence of
bisphosphonates alendronic acid (Ringe et al. 2002), sufficient androgen and/or estrogen concentrations
risedronic acid (Boonen et al. 2009) and zoledronic which require androgen and estrogen receptor alpha
acid (Smith et al. 2003), as well the RANKL (receptor (not beta) respectively. Cortical bone medullary
activator of nuclear factor-kB ligand) antagonist expansion also increases in the elderly rodent, which
denosumab (Smith et al. 2009), that have a well-docu- is insufficiently counterbalanced by periosteal bone
mented anti-fracture efficacy in postmenopausal formation. Estrogen inhibits net endocortical bone
osteoporosis, also increase bone density in osteoporo- resorption, and testosterone may stimulate periosteal
tic male patients with concomitant low testosterone bone formation but its effect is weaker compared to
levels. Two-year treatment with zoledronic acid also younger bones (Vanderschueren et al. 2004).
decreased vertebral fracture risk overall in the patient An important finding from animal studies is that
group with osteoporosis (Brown et al. 2007). the effect of testosterone on bone may occur at sub-
An anabolic agent such as parathyroid hormone physiological concentrations in these elderly non-
(PTH), approved for treatment of osteoporosis in post- growing rodents (Vanderschueren et al. 2000). In a
menopausal women, also increases bone density to a comparative study, the effect of non-aromatizable
similar extent in men and women (Misiorowski 2010). androgens is significant but less potent than testoster-
Therefore, physicians currently have a choice one in this animal model (Vandenput et al. 2002).
between different treatment modalities for efficient If translated to humans, this preclinical observation
prevention of fracture risk, even in hypogonadal may explain why extensive and rapid bone loss at both
patients not receiving testosterone replacement. cortical and trabecular sites is only observed when tes-
tosterone concentrations are very low, as is the case after
8.8 Preclinical studies of sex steroid orchidectomy. Bone loss observed in elderly men with
normal and/or borderline low testosterone is indeed
replacement in male rodents much slower than the rapid bone loss observed after
As discussed, the relative contribution of non-aroma- orchidectomy. Bone loss in aging men is also mainly
tizable versus aromatizable androgen action on bone caused by trabecular thinning and less by trabecular
development and maintenance remains not well disruption, as observed following castration. Therefore,
defined in humans. However, in male orchidecto- the mechanism of age-related bone loss in the human is
mized rats, both non-aromatizable (Venken et al. clearly different from the bone loss observed after orch-
2005) and aromatizable androgens (Vanderschueren idectomy in the rodent. Lower estrogen concentrations
et al. 2000) increase bone formation during skeletal in aging men may, even in the presence of normal
growth and decrease bone resorption after growth. testosterone concentrations, enhance endosteal
During growth, testosterone increases cortical bone bone resorption (Falahati-Nini et al. 2000; Van-
size (periosteal bone formation) and length (longitu- derschueren et al. 2004; Reim et al. 2008). The relative
dinal bone formation). These actions are partly related and potentially important role of estrogen in the main-
to a direct action of androgens via the AR and partly to tenance of human bone is, however, much more diffi-
an indirect action of androgens through aromatization cult to address in preclinical models because of lower

183
Chapter 8: Testosterone and bone

serum estrogen concentration in rodents compared to Sex hormone


men (Falahati-Nini et al. 2000).

8.9 Potentialmechanismofandrogen Osteoblast

receptor-mediated androgen action


RANKL
Three major types of mature bone cells are present OPG
within the bone matrix: osteocytes, osteoblasts and RANK
osteoclasts.
Osteoblasts are responsible for the synthesis of Osteoclast c-fms M-CSF
matrix constituents and bone formation, and arise precursor
from multipotential mesenchymal stem cells (Mano-
lagas 2000; Harada and Rodan 2003). The osteodiffer-
entiation originates from a progenitor cell, the
osteoprogenitor, and progresses through a number
of precursor stages to the mature osteoblast (Harada Mature
and Rodan 2003). In this regard, an essential event in osteoclast
osteoblast differentiation is the activation of runt-
related transcription factor 2 (Runx2) and osterix
(Osx), as mice deficient in one of these transcription BONE
factors completely lack osteoblasts (Ducy 2000; Har- Fig. 8.1 A schematic overview of the RANKL/RANK/OPG system.
ada and Rodan 2003). Receptor activator of nuclear factor-kB ligand mediates a signal for
osteoclast formation through RANK expressed on osteoclast
Osteocytes are terminally differentiated osteo- progenitors. Osteoprotegerin counteracts this effect by competing
blasts that are fully embedded in the secreted matrix for and neutralizing RANKL. Osteoclast differentiation factor is a
(Bonewald and Johnson 2008). ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor.
M-CSF is an essential factor for osteoclast proliferation and
Osteoclasts, the exclusive bone resorbing cells, are differentiation, which is produced by osteoblasts in osseous tissues
multinucleated cells that arise from hematopoietic and acts through its receptor c-fms. See plate section for color
stem cells of the monocyte/macrophage lineage (Man- version.
olagas 2000; Boyle et al. 2003). Two cytokines are
essential for the commitment of hematopoietic stem
cells to the myeloid lineage: receptor activator of demonstrated that osteoclasts express ARs and that
nuclear factor-kB ligand (RANKL) and macrophage- DHT directly inhibits the resorptive capacity of iso-
colony stimulating factor (M-CSF). These proteins lated human, murine and avian osteoclasts in vitro.
are produced by marrow stromal cells and osteoblasts. Furthermore, androgens may also directly modulate
Binding of RANKL to its receptor RANK, expressed RANKL-induced osteoclast formation, independently
on osteoclasts and its hematopoietic precursor cells, is of bone marrow cells (Fig. 8.1) (Huber et al. 2001). The
necessary for osteoclast formation and differentiation. impact of AR-mediated androgen action was illus-
These stimulating effects on osteoclastogenesis are trated by the study of Kawano et al., who showed that
counterbalanced by osteoprotegerin (OPG), which is the suppressive function of the AR system on RANKL
secreted by stromal cells and osteoblasts, by acting as gene expression mediates the protective effects of
a soluble inhibitor of RANKL (Boyle et al. 2003). androgens on bone remodeling through the inhibition
Androgens may inhibit bone resorption, but the of bone resorption (Kawano et al. 2003). Analysis of
mechanism of AR-mediated action on bone cells is primary osteoblasts and osteoclasts from global AR
not well established. One of the open questions is knock-out mice indeed revealed that AR function was
which bone cell constitutes the principal target for required for the suppressive effects of androgens on
androgen action. Indeed, both osteoblasts and osteo- osteoclastogenesis and that this AR function was medi-
clasts, as well as their respective precursor cells, may ated by osteoblasts (Kawano et al. 2003).
be a target for androgen action. Besides the RANKLOPG pathway, AR-mediated
Androgens may have a direct effect on the preos- action may be involved in other aspects of function
teoclasts/osteoclasts. Pederson et al. (1999) have and interaction of osteoblasts and osteoclasts.

184
Chapter 8: Testosterone and bone

It is well established that estrogens affect gener- 8.10 Key messages


ation and survival of osteoblasts and osteoclasts.  Hypogonadism and therefore sex steroid
According to Nakamura et al. (2007), estrogens deficiency is a well-established cause of
reduce number and survival of osteoclasts, and there- high turnover osteoporosis, but most
fore bone resorption, in female but not in male mice, osteoporotic male patients do not suffer from
hereby again suggesting that the AR function may be hypogonadism.
more important in the male. Possibly, the AR also acts  Testosterone replacement maintains bone in
via Runx2, an osteoblast master transcription factor hypogonadal men, but its anti-fracture efficacy is
which promotes osteoblast-mediated osteoclastogen- not well documented.
esis and bone resorption via stimulation of genes  The extent to which the effect of testosterone
replacement in men is due to either AR-mediated
which encode for M-CSF and RANKL. The binding
action or to conversion of estrogens is not fully
of the AR to Runx2 inhibits Runx2 functioning in understood.
osteoblasts during later stages of their development.  A dual mode of action of testosterone is,
In mice, this AR-mediated repression of Runx2 in however, clearly established in animal model
osteoblasts resulted in restrained osteoclastogenesis studies, but the underlying mechanism of the
(Baniwal et al. 2009). Accumulation of ROS during aformentioned actions and bone target cells are
the aging process was also suggested as a potential still unclear.
mechanism of senile as well as hypogonadal osteopor-  Both androgens and estrogens may have an
osis: ROS indeed appear to influence both generation effect on osteoclast and osteoblast generation
and survival of osteoclasts, osteoblasts and osteocytes. and survival as well as cross-talk between
osteoblasts and osteoclasts.
Declining levels of estrogens or androgens decrease
 Many of the agents used for the treatment of
the defense mechanisms against oxidative stress in
postmenopausal women also appear to be
bone, and this may account for the increased bone effective in men suffering from osteoporosis.
resorption associated with the acute loss of both of
these hormones (Manolagas 2010).

8.11 References estradiol level is positively related to


muscle strength and physical
Schneider DL, Sartoris DJ (2000)
Low levels of estradiol are associated
Amin S, Zhang Y, Felson DT, Sawin performance independent of muscle with vertebral fractures in older
CT, Hannan MT, Wilson PW, Kiel mass: a cross-sectional study in men, but not women: the Rancho
DP (2006) Estradiol, testosterone, 1489 older men. Eur J Endocrinol Bernardo Study. J Clin Endocrinol
and the risk for hip fractures in 164:811817 Metab 85:219223
elderly men from the Framingham
Study. Am J Med 119:426433 Baim S, Leonard MB, Bianchi ML, Basaria S, Dobs AS (2001)
Hans DB, Kalkwarf HJ, Langman Hypogonadism and androgen
Anderson FH, Francis RM, Peaston
CB, Rauch F (2008) Official replacement therapy in elderly men.
RT, Wastell HJ (1997) Androgen
positions of the International Am J Med 110:563572
supplementation in eugonadal men
Society for Clinical Densitometry Behre HM, Kliesch S, Leifke E, Link
with osteoporosis: effects of six
and executive summary of the 2007 TM, Nieschlag E (1997) Long-term
months treatment on markers of
ISCD Pediatric Position effect of testosterone therapy on
bone formation and resorption.
Development Conference. J Clin
J Bone Miner Res 12:472478 bone mineral density in
Densitom 11:621 hypogonadal men. J Clin Endocrinol
Araujo AB, Travison TG, Leder BZ,
Baniwal SK, Khalid O, Sir D, Buchanan Metab 82:23862390
Mckinlay JB (2008) Correlations
between serum testosterone, G, Coetzee GA, Frenkel B (2009) Bertelloni S, Baroncelli GI, Battini R,
estradiol, and sex hormone- Repression of Runx2 by androgen Perri G, Saggese G (1995) Short-
binding globulin and bone mineral receptor (AR) in osteoblasts and term effect of testosterone treatment
density in a diverse sample of men. prostate cancer cells: AR binds on reduced bone density in boys
J Clin Endocrinol Metab 93:2135 Runx2 and abrogates its with constitutional delay of puberty.
2141 recruitment to DNA. Mol J Bone Miner Res 10:14881495
Endocrinol 23:12031214
Auyeung TW, Lee JS, Kwok T, Leung J, Bertelloni S, Baroncelli GI, Ferdeghini
Ohlsson C, Vandenput L, Leung PC, Barrett-Connor E, Mueller JE, Von M, Perri G, Saggese G (1998)
Woo J (2011) Testosterone but not Muhlen DG, Laughlin GA, Normal volumetric bone mineral

185
Chapter 8: Testosterone and bone

density and bone turnover in young in older men with low testosterone De Laet CE, Van Hout BA, Burger H,
men with histories of constitutional levels. J Clin Endocrinol Metab Weel AE, Hofman A, Pols HA
delay of puberty. J Clin Endocrinol 94:47854792 (1998) Hip fracture prediction in
Metab 83:42804283 Callewaert F, Venken K, Ophoff J, De elderly men and women: validation
Bilezikian JP, Morishima A, Bell J, Gendt K, Torcasio A, Van Lenthe in the Rotterdam study. J Bone
Grumbach MM (1998) Increased GH, Van Oosterwyck H, Boonen S, Miner Res 13:15871593
bone mass as a result of estrogen Bouillon R, Verhoeven G, Doran PM, Riggs BL, Atkinson EJ,
therapy in a man with aromatase Vanderschueren D (2009) Khosla S (2001) Effects of raloxifene,
deficiency. N Engl J Med 339:599603 Differential regulation of bone and a selective estrogen receptor
Bjornerem A, Ahmed LA, Joakimsen body composition in male mice modulator, on bone turnover
RM, Berntsen GK, Fonnebo V, with combined inactivation of markers and serum sex steroid and
Jorgensen L, Oian P, Seeman E, androgen and estrogen receptor- lipid levels in elderly men. J Bone
Straume B (2007) A prospective alpha. FASEB J 23:232240 Miner Res 16:21182125
study of sex steroids, sex hormone- Callewaert F, Boonen S, Ducy P (2000) Cbfa1: a molecular
binding globulin, and non-vertebral Vanderschueren D (2010a) Sex switch in osteoblast biology. Dev
fractures in women and men: the steroids and the male skeleton: a tale Dyn 219:461471
Tromso Study. Eur J Endocrinol of two hormones. Trends Endocrinol Dunkel L, Wickman S (2002) Novel
157:119125 Metab 21:8995 treatment of delayed male puberty
Bonewald LF, Johnson ML (2008) Callewaert F, Sinnesael M, Gielen E, with aromatase inhibitors. Horm
Osteocytes, mechanosensing and Boonen S, Vanderschueren D Res 57 (Suppl 2):4452
Wnt signaling. Bone 42:606615 (2010b) Skeletal sexual dimorphism: Empana JP, Dargent-Molina P,
Boonen S, Vanderschueren D, relative contribution of sex steroids, Breart G (2004) Effect of hip
Geusens P, Bouillon R (1997) Age- GH-IGF1, and mechanical loading. fracture on mortality in elderly
associated endocrine deficiencies as J Endocrinol 207:127134 women: the EPIDOS prospective
potential determinants of femoral Carani C, Qin K, Simoni M, Faustini- study. J Am Geriatr Soc 52:685690
neck (type II) osteoporotic fracture Fustini M, Serpente S, Boyd J, Ensrud KE, Thompson DE, Cauley JA,
occurrence in elderly men. Int Korach KS, Simpson ER (1997) Nevitt MC, Kado DM, Hochberg
J Androl 20:134143 Effect of testosterone and estradiol MC, Santora AC 2nd, Black DM
Boonen S, Orwoll ES, Wenderoth D, in a man with aromatase deficiency. (2000) Prevalent vertebral
Stoner KJ, Eusebio R, Delmas PD N Engl J Med 337:9195 deformities predict mortality and
(2009) Once-weekly risedronate in Center JR, Nguyen TV, Schneider D, hospitalization in older women with
men with osteoporosis: results of a Sambrook PN, Eisman JA (1999) low bone mass. Fracture
2-year, placebo-controlled, double- Mortality after all major types of Intervention Trial Research Group.
blind, multicenter study. J Bone osteoporotic fracture in men and J Am Geriatr Soc 48:241249
Miner Res 24:719725 women: an observational study. Fabbri A, Giannetta E, Lenzi A, Isidori
Bouillon R, Bex M, Vanderschueren D, Lancet 353:878882 AM (2007) Testosterone treatment
Boonen S (2004) Estrogens are Crawford BA, Liu PY, Kean MT, to mimic hormone physiology in
essential for male pubertal Bleasel JF, Handelsman DJ (2003) androgen replacement therapy.
periosteal bone expansion. J Clin Randomized placebo-controlled A view on testosterone gel and other
Endocrinol Metab 89:60256029 trial of androgen effects on muscle preparations available. Expert Opin
Boyle WJ, Simonet WS, Lacey DL and bone in men requiring long- Biol Ther 7:10931106
(2003) Osteoclast differentiation term systemic glucocorticoid Falahati-Nini A, Riggs BL, Atkinson EJ,
and activation. Nature 423:337342 treatment. J Clin Endocrinol Metab Ofallon WM, Eastell R, Khosla S
88:31673176 (2000) Relative contributions of
Brown JE, Ellis SP, Lester JE,
Gutcher S, Khanna T, Purohit OP, Daniell HW (1997) Osteoporosis after testosterone and estrogen in
Mccloskey E, Coleman RE (2007) orchiectomy for prostate cancer. regulating bone resorption and
Prolonged efficacy of a single dose J Urol 157:439444 formation in normal elderly men.
of the bisphosphonate zoledronic De Laet CE, Van Hout BA, Burger H, J Clin Invest 106:15531560
acid. Clin Cancer Res 13:54065410 Hofman A, Pols HA (1997) Fechtenbaum J, Cropet C, Kolta S,
Burnett-Bowie SA, Mckay EA, Lee H, Bone density and risk of hip Horlait S, Orcel P, Roux C (2005)
Leder BZ (2009) Effects of fracture in men and women: The severity of vertebral fractures
aromatase inhibition on bone cross sectional analysis. BMJ and health-related quality of life in
mineral density and bone turnover 315:221225 osteoporotic postmenopausal

186
Chapter 8: Testosterone and bone

women. Osteoporos Int Harada S, Rodan GA (2003) Control of Kanis JA, Johnell O, Oden A, Sembo I,
16:21752179 osteoblast function and regulation Redlund-Johnell I, Dawson A, De
Ferlin A, Schipilliti M, Vinanzi C, of bone mass. Nature Laet C, Jonsson B (2000) Long-term
Garolla A, Di Mambro A, Selice R, 423:349355 risk of osteoporotic fracture in
Lenzi A, Foresta C (2011) Bone Holt G, Khaw KT, Reid DM, Malmo. Osteoporos Int 11:669674
mass in subjects with Klinefelter Compston JE, Bhalla A, Woolf AD, Kanis JA, Johnell O, Oden A,
syndrome: role of testosterone levels Crabtree NJ, Dalzell N, Wardley- Johansson H, Mccloskey E (2008)
and androgen receptor gene CAG Smith B, Lunt M, Reeve J (2002) FRAX and the assessment of
polymorphism. J Clin Endocrinol Prevalence of osteoporotic bone fracture probability in men and
Metab 96:E739E745 mineral density at the hip in Britain women from the UK. Osteoporos Int
Finkelstein JS, Klibanski A, Neer RM, differs substantially from the US 19:385397
Greenspan SL, Rosenthal DI, over 50 years of age: implications
Katznelson L, Finkelstein JS,
Crowley WF Jr (1987) Osteoporosis for clinical densitometry. Br J Radiol
Schoenfeld DA, Rosenthal DI,
in men with idiopathic 75:736742
Anderson EJ, Klibanski A (1996)
hypogonadotropic hypogonadism. Huber DM, Bendixen AC, Pathrose P, Increase in bone density and lean
Ann Intern Med 106:354361 Srivastava S, Dienger KM, Shevde body mass during testosterone
Finkelstein JS, Neer RM, Biller BM, NK, Pike JW (2001) Androgens administration in men with
Crawford JD, Klibanski A (1992) suppress osteoclast formation acquired hypogonadism. J Clin
Osteopenia in men with a history of induced by RANKL and Endocrinol Metab 81:43584365
delayed puberty. N Engl J Med macrophage-colony stimulating
Kawano H, Sato T, Yamada T,
326:600604 factor. Endocrinology
Matsumoto T, Sekine K, Watanabe T,
142:38003808
Finkelstein JS, Klibanski A, Neer RM Nakamura T, Fukuda T, Yoshimura K,
Isidori AM, Giannetta E, Greco EA, Yoshizawa T, Aihara K, Yamamoto Y,
(1999) Evaluation of lumber spine
Gianfrilli D, Bonifacio V, Isidori A, Nakamichi Y, Metzger D, Chambon P,
bone mineral density (BMD) using
Lenzi A, Fabbri A (2005) Effects of Nakamura K, Kawaguchi H, Kato S
dual energy X-ray absorptiometry
testosterone on body composition, (2003) Suppressive function of
(DXA) in 21 young men with
bone metabolism and serum lipid androgen receptor in bone
histories of constitutionally-delayed
profile in middle-aged men: a meta- resorption. Proceedings of the
puberty. J Clin Endocrinol
analysis. Clin Endocrinol (Oxf) National Academy of Sciences of
Metab 84:34003401; author
63:280293 the United States of America
reply:34033404
Jackson JA, Kleerekoper M, Parfitt AM, 100:94169421
Fried LP, Tangen CM, Walston J, Rao DS, Villanueva AR, Frame B
Newman AB, Hirsch C, Kearbey JD, Gao W, Fisher SJ, Wu D,
(1987) Bone histomorphometry in Miller DD, Dalton JT (2009) Effects
Gottdiener J, Seeman T, Tracy R, hypogonadal and eugonadal men
Kop WJ, Burke G, Mcburnie MA of selective androgen receptor
with spinal osteoporosis. J Clin modulator (SARM) treatment in
(2001) Frailty in older adults: Endocrinol Metab 65:5358
evidence for a phenotype. J Gerontol osteopenic female rats. Pharm Res
A Biol Sci Med Sci 56:M146M156 Johnell O, Kanis JA (2006) An estimate 26:24712477
of the worldwide prevalence and Kelepouris N, Harper KD, Gannon F,
Gao W, Reiser PJ, Coss CC, Phelps disability associated with Kaplan FS, Haddad JG (1995)
MA, Kearbey JD, Miller DD, Dalton osteoporotic fractures. Osteoporos Severe osteoporosis in men. Ann
JT (2005) Selective androgen Int 17:17261733 Intern Med 123:452460
receptor modulator treatment
improves muscle strength and body Kamel HK (2005) Male osteoporosis: Kenny M, Prestwood KM, Gruman
composition and prevents bone loss new trends in diagnosis and CA, Marcello KM, Raisz LG (2001)
in orchidectomized rats. therapy. Drugs Aging 22:741748 Effects of transdermal testosterone
Endocrinology 146:48874897 Kanis JA (2002) Diagnosis of on bone and muscle in older men
osteoporosis and assessment of with low bioavailable testosterone
Goderie-Plomp HW, Van Der Klift M,
fracture risk. Lancet 359:19291936 levels. J Gerontol A Biol Sci Med Sci
De Ronde W, Hofman A, De Jong
Kanis JA, Gluer CC (2000) An update 56:M266M272
FH, Pols HA (2004) Endogenous
sex hormones, sex hormone- on the diagnosis and assessment of Kenny M, Prestwood KM, Gruman
binding globulin, and the risk of osteoporosis with densitometry. CA, Fabregas G, Biskup B,
incident vertebral fractures in Committee of Scientific Advisors, Mansoor G (2002) Effects of
elderly men and women: the International Osteoporosis transdermal testosterone on lipids
Rotterdam Study. J Clin Endocrinol Foundation. Osteoporos Int and vascular reactivity in older
Metab 89:32613269 11:192202 men with low bioavailable

187
Chapter 8: Testosterone and bone

testosterone levels. J Gerontol A Biol the pathogenesis and treatment of mutation and the physiological role
Sci Med Sci 57:M460M465 osteoporosis. Endocr Rev of estrogens. J Clin Endocrinol
Khosla S, Lufkin EG, Hodgson SF, 21:115137 Metab 80:36893698
Fitzpatrick LA, Melton LJ 3rd Manolagas SC (2010) From estrogen- Nakamura T, Imai Y, Matsumoto T,
(1994) Epidemiology and clinical centric to aging and oxidative stress: Sato S, Takeuchi K, Igarashi K,
features of osteoporosis in young a revised perspective of the Harada Y, Azuma Y, Krust A,
individuals. Bone 15:551555 pathogenesis of osteoporosis. Yamamoto Y, Nishina H, Takeda S,
Khosla S, Melton LJ 3rd, Atkinson EJ, Endocr Rev 31:266300 Takayanagi H, Metzger D, Kanno J,
Ofallon WM, Klee GG, Riggs BL Maravic M, Le Bihan C, Landais P, Takaoka K, Martin TJ, Chambon P,
(1998) Relationship of serum sex Fardellone P (2005) Incidence and Kato S (2007) Estrogen prevents
steroid levels and bone turnover cost of osteoporotic fractures in bone loss via estrogen receptor
markers with bone mineral density France during 2001. alpha and induction of Fas ligand in
in men and women: a key role for A methodological approach by the osteoclasts. Cell 130:811823
bioavailable estrogen. J Clin national hospital database. OConnell M, Tajar A, Roberts S, Wu F
Endocrinol Metab 83:22662274 Osteoporos Int 16:14751480 (2011) Do androgens play any role
Khosla S, Amin S, Orwoll E (2008) Meier C, Nguyen TV, Handelsman DJ, in the physical frailty of ageing
Osteoporosis in men. Endocr Rev Schindler C, Kushnir MM, men? J Androl 34:17
29:441464 Rockwood AL, Meikle W, Center Orsini LS, Rousculp MD, Long SR,
Kurland ES, Rosen CJ, Cosman F, JR, Eisman JA, Seibel MJ (2008) Wang S (2005) Health care
Mcmahon D, Chan F, Shane E, Endogenous sex hormones and utilization and expenditures in the
Lindsay R, Dempster D, Bilezikian incident fracture risk in older men: United States: a study of
JP (1997) Insulin-like growth the Dubbo Osteoporosis osteoporosis-related fractures.
factor-I in men with idiopathic Epidemiology Study. Arch Intern Osteoporos Int 16:359371
osteoporosis. J Clin Endocrinol Med 168:4754 Page ST, Amory JK, Bowman FD,
Metab 82:27992805 Meier DE, Orwoll ES, Keenan EJ, Anawalt BD, Matsumoto M,
Lanfranco F, Zirilli L, Baldi M, Fagerstrom RM (1987) Marked Bremner WJ, Tenover JL (2005)
Pignatti E, Corneli G, Ghigo E, decline in trabecular bone mineral Exogenous testosterone (T) alone or
Aimaretti G, Carani C, Rochira V content in healthy men with age: with finasteride increases physical
(2008) A novel mutation in the lack of association with sex steroid performance, grip strength, and
human aromatase gene: insights on levels. J Am Geriatr Soc 35:189197 lean body mass in older men with
the relationship among serum Mellstrom D, Vandenput L, Mallmin low serum T. J Clin Endocrinol
estradiol, longitudinal growth and H, Holmberg AH, Lorentzon M, Metab 90:15021510
bone mineral density in an adult Oden A, Johansson H, Orwoll ES, Pederson L, Kremer M, Judd J,
man under estrogen replacement Labrie F, Karlsson MK, Pascoe D, Spelsberg TC, Riggs BL,
treatment. Bone 43:628635 Ljunggren O, Ohlsson C (2008) Oursler MJ (1999) Androgens
Lippuner K, Golder M, Greiner R Older men with low serum estradiol regulate bone resorption activity of
(2005) Epidemiology and direct and high serum SHBG have an isolated osteoclasts in vitro. Proc
medical costs of osteoporotic increased risk of fractures. J Bone Natl Acad Sci USA 96:505510
fractures in men and women in Miner Res 23:15521560 Pelat C, Van Pottelbergh I, Cohen-Solal
Switzerland. Osteoporos Int 16 Melton LJ 3rd, Lieber MM, Atkinson M, Ostertag A, Kaufman JM,
Suppl 2:S8S17 EJ, Achenbach SJ, Zincke H, Martinez M, De Vernejoul MC
Ly LP, Jimenez M, Zhuang TN, Therneau TM, Khosla S (2011) (2007) Complex segregation
Celermajer DS, Conway J, Fracture risk in men with prostate analysis accounting for GxE of bone
Handelsman DJ (2001) A double- cancer: a population-based study. mineral density in European
blind, placebo-controlled, J Bone Miner Res 26:18081815 pedigrees selected through a male
randomized clinical trial of Misiorowski W (2010) Parathyroid proband with low BMD. Ann Hum
transdermal dihydrotestosterone gel hormone and its analogues Genet 71:2942
on muscular strength, mobility, and molecular mechanisms of action Pernow Y, Granberg B, Saaf M,
quality of life in older men with and efficacy in osteoporosis therapy. Weidenhielm L (2006) Osteoblast
partial androgen deficiency. J Clin Endokrynol Pol 62:7378 dysfunction in male idiopathic
Endocrinol Metab 86:40784088 Morishima A, Grumbach MM, osteoporosis. Calcif Tissue Int
Manolagas SC (2000) Birth and death Simpson ER, Fisher C, Qin K (1995) 78:9097
of bone cells: basic regulatory Aromatase deficiency in male and Pernow Y, Hauge EM, Linder K,
mechanisms and implications for female siblings caused by a novel Dahl E, Saaf M (2009) Bone

188
Chapter 8: Testosterone and bone

histomorphometry in male Seeman E (2001) Clinical review 137: years of age. J Clin Endocrinol
idiopathic osteoporosis. Calcif Sexual dimorphism in skeletal size, Metab 84:19661972
Tissue Int 84:430438 density, and strength. J Clin Stepan JJ, Lachman M, Zverina J,
Rapado A, Hawkins F, Sobrinho L, Endocrinol Metab 86: Pacovsky V, Baylink DJ (1989)
Diaz-Curiel M, Galvao-Telles A, 45764584 Castrated men exhibit bone loss:
Arver S, Melo Gomes J, Mazer N, Seeman E (2003) Periosteal bone effect of calcitonin treatment on
Garcia E, Costa J, Horcajada C, formation a neglected biochemical indices of bone
Lopez-Gavilanes E, Mascarenhas M, determinant of bone strength. remodeling. J Clin Endocrinol Metab
Papapietro K, Lopez Alvarez MB, N Engl J Med 349:320323 69:523527
Pereira MC, Martinez G, Valverde I, Smith EP, Boyd J, Frank GR, Takahashi Taes Y, Lapauw B, Vandewalle S,
Garcia JJ, Carballal JJ, Garcia I H, Cohen RM, Specker B, Williams Zmierczak H, Goemaere S,
(1999) Bone mineral density and TC, Lubahn DB, Korach KS (1994) Vanderschueren D, Kaufman JM,
androgen levels in elderly males. Estrogen resistance caused by a Tsjoen G (2009) Estrogen-specific
Calcif Tissue Int 65:417421 mutation in the estrogen-receptor action on bone geometry and
Rariy CM, Ratcliffe SJ, Weinstein R, gene in a man. N Engl J Med volumetric bone density:
Bhasin S, Blackman MR, Cauley JA, 331:10561061 longitudinal observations in an
Robbins J, Zmuda JM, Harris TB, adult with complete androgen
Smith EP, Specker B, Bachrach BE,
Cappola R (2011) Higher serum free Kimbro KS, Li XJ, Young MF, insensitivity. Bone 45:392397
testosterone concentration in older
Fedarko NS, Abuzzahab MJ, Frank Vandenput L, Boonen S, Van Herck E,
women is associated with greater GR, Cohen RM, Lubahn DB, Swinnen JV, Bouillon R,
bone mineral density, lean body
Korach KS (2008) Impact on bone Vanderschueren D (2002) Evidence
mass, and total fat mass: the of an estrogen receptor-alpha gene from the aged orchidectomized
cardiovascular health study. J Clin
loss of function mutation. J Clin male rat model that 17beta-estradiol
Endocrinol Metab 96:989996 Endocrinol Metab 93:30883096 is a more effective bone-sparing and
Reid IR (1998) Glucocorticoid-induced anabolic agent than 5alpha-
Smith MR, Eastham J, Gleason DM,
osteoporosis: assessment and dihydrotestosterone. J Bone Miner
Shasha D, Tchekmedyian S,
treatment. J Clin Densitom 1:6573 Res 17:20802086
Zinner N (2003) Randomized
Reifenstein EC Jr, Albright F (1947) controlled trial of zoledronic acid to Vanderschueren D, Vandenput L,
The metabolic effects of steroid prevent bone loss in men receiving Boonen S, Van Herck E, Swinnen
hormones in osteoporosis. J Clin androgen deprivation therapy for JV, Bouillon R (2000) An aged rat
Invest 26:2456 nonmetastatic prostate cancer. model of partial androgen
Reim NS, Breig B, Stahr K, Eberle J, J Urol 169:20082012 deficiency: prevention of both loss
Hoeflich A, Wolf E, Erben RG Smith MR, Fallon MA, Lee H, of bone and lean body mass by low-
(2008) Cortical bone loss in Finkelstein JS (2004) Raloxifene to dose androgen replacement.
androgen-deficient aged male rats is prevent gonadotropin-releasing Endocrinology 141:16421647
mainly caused by increased hormone agonist-induced bone loss Vanderschueren D, Vandenput L,
endocortical bone remodeling. in men with prostate cancer: a Boonen S, Lindberg MK,
J Bone Miner Res 23:694704 randomized controlled trial. J Clin Bouillon R, Ohlsson C (2004)
Ringe JD, Orwoll E, Daifotis A, Endocrinol Metab 89:38413846 Androgens and bone. Endocr Rev
Lombardi A (2002) Treatment of Smith MR, Egerdie B, Hernandez Toriz 25:389425
male osteoporosis: recent advances N, Feldman R, Tammela TL, Saad F, Vanderschueren D, Vandenput L,
with alendronate. Osteoporos Int Heracek J, Szwedowski M, Ke C, Boonen S (2005) Reversing sex
13:195199 Kupic A, Leder BZ, Goessl C (2009) steroid deficiency and optimizing
Rockwood K, Mitnitski A (2007) Denosumab in men receiving skeletal development in the
Frailty in relation to the androgen-deprivation therapy for adolescent with gonadal failure.
accumulation of deficits. J Gerontol prostate cancer. N Engl J Med Endocr Dev 8:150165
A Biol Sci Med Sci 62:722727 361:745755 Venken K, Boonen S, Van Herck E,
Schatzl G, Madersbacher S, Temml C, Snyder PJ, Peachey H, Hannoush P, Vandenput L, Kumar N, Sitruk-
Krenn-Schinkel K, Nader A, Berlin JA, Loh L, Holmes JH, Ware R, Sundaram K, Bouillon R,
Sregi G, Lapin A, Hermann M, Dlewati A, Staley J, Santanna J, Vanderschueren D (2005) Bone and
Berger P, Marberger M (2003) Kapoor SC, Attie MF, Haddad JG Jr, muscle protective potential of the
Serum androgen levels in men: Strom BL (1999) Effect of prostate-sparing synthetic androgen
impact of health status and age. testosterone treatment on bone 7alpha-methyl-19-nortestosterone:
Urology 61:629633 mineral density in men over 65 evidence from the aged

189
Chapter 8: Testosterone and bone

orchidectomized male rat model. growing male mice: a study in the (1999) Androgen deprivation
Bone 36:663670 androgen receptor knockout mouse therapy for prostate cancer results
Venken K, De Gendt K, Boonen S, model. J Bone Miner Res 21:576585 in significant loss of bone density.
Ophoff J, Bouillon R, Swinnen JV, Urology 54:607611
Vermeulen A, Kaufman JM, Goemaere
Verhoeven G, Vanderschueren D S, Van Pottelberg I (2002) Estradiol Zirilli L, Rochira V, Diazzi C, Caffagni
(2006) Relative impact of androgen in elderly men. Aging Male 5:98102 G, Carani C (2008) Human
and estrogen receptor activation in models of aromatase deficiency.
the effects of androgens on Wei JT, Gross M, Jaffe CA, Gravlin K, J Steroid Biochem Mol Biol 109:
trabecular and cortical bone in Lahaie M, Faerber GJ, Cooney KA 212218

190
Chapter
Androgen effects on the skeletal muscle

9 Shalender Bhasin, Ravi Jasuja, Carlo Serra, Rajan Singh, Thomas W. Storer,
Wen Guo, Thomas G. Travison, and Shezad Basaria

9.1 Introduction 191 9.8 Testosterones effects on mood, fatigue/


9.2 The association of circulating testosterone energy, sense of well-being and quality of
concentrations and muscle mass, muscle life 196
performance and physical function: 9.9 Reaction time 197
epidemiological evidence 192 9.10 Strategies for achieving selectivity of
9.3 The effects of experimental lowering of testosterone action: dissociating the
endogenous testosterone concentrations on beneficial effects of testosterone from its
body composition 192 adverse effects 197
9.4 The effects of physiological testosterone 9.10.1 Mechanisms by which testosterone
replacement in models of androgen increases skeletal muscle mass 197
deficiency 192 9.10.2 The role of circulating growth hormone
9.5 Testosterone dose-response relationships in and IGF-1, and intramuscular IGF-1
young and older men 193 signaling in mediating testosterones
9.6 Randomized controlled trials of testosterone effects on the muscle 199
and other androgens in men with chronic 9.11 Perspective on the future of testosterone as a
diseases 193 function-promoting therapy 200
9.7 Randomized trials of testosterone in healthy 9.12 Key messages 200
older men 194 9.13 References 200

9.1 Introduction with an intent to produce more selective androgens


The idea that androgens have anabolic effects on the with enhanced anabolic activity. These androgenic-
muscle is not novel; humans have known since anabolic steroids were first used in professional
antiquity that the removal of the testes depletes mens sports by Russian weightlifters; however, the use of
vigor. In modern times, shortly after the synthesis of androgens spread widely to athletes in other parts of
testosterone, Kochakian at the University of Roches- the world and to other sports (Yesalis et al. 1993;
ter and Kenyon et al. at the University of Chicago Yesalis and Bahrke 1995; Freeman et al. 2001). In
provided unequivocal evidence of the anabolic effects the early 1980s, with the availability of underground
of testosterone by demonstrating that androgen steroid handbooks, the use of androgens spilled over
administration increases nitrogen retention in cas- from the athletes into the general community of
trated males of many mammalian species, and in recreational bodybuilders. In spite of the wide
eunuchoidal men and women (Kochakian and Murlin empiric evidence of anabolic activity of androgens
1935; Kochakian 1937; 1950; Kenyon et al. 1940). The emerging from the experience of athletes and body-
decades of 1940 and 1950 witnessed the synthesis of builders, the academic community continued to
novel steroidal androgen receptor modulators, mostly maintain a puritanical denial; the Endocrine Society

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

191
Chapter 9: Androgen effects on the skeletal muscle

issued a position statement declaring that anabolic Beld et al. 2000; Schaap et al. 2005; Orwoll et al. 2006;
steroids do not enhance muscle mass or strength. In Krasnoff et al. 2009; Cawthon et al. 2009). In the Osteo-
the mid-1990s, the publication of well-conducted, porotic Fractures in Men Study (MrOS), low bioavail-
randomized clinical trials finally put this controversy able testosterone levels were associated with lower levels
to rest, demonstrating unequivocally that when of physical performance and increased risk of falls
energy and protein intake, and exercise stimulus are (Orwoll et al. 2006). The association of bioavailable
standardized, the administration of a supraphysiolo- testosterone level with fall risk in the MrOS study per-
gical dose of testosterone increases fat-free mass and sisted even after adjusting for measures of physical per-
maximal voluntary strength, and that the concomi- formance, suggesting that testosterone levels may reduce
tant administration of a standardized resistance exer- fall propensity through other mechanisms (Orwoll et al.
cise program augments the anabolic effects of 2006). Low free testosterone levels have also been associ-
androgens on muscle mass and maximal voluntary ated with increased risk of incident mobility limitations
strength (Bhasin et al. 1996). Since then, substantial and progression of mobility limitation in men in the
pharmaceutical and academic research effort has Framingham Heart Study (Krasnoff et al. 2009).
focused on the development of androgens as func- Epidemiological studies have demonstrated an
tion-promoting anabolic therapies. This chapter will inverse relationship between serum testosterone levels
review the large body of epidemiological and clinical and measures of obesity and visceral fat mass (Seidell
trial evidence of the anabolic effects of androgens et al. 1990; Khaw and Barrett-Connor 1992). Total and
on skeletal muscle, and explore the hypothesis that free testosterone concentrations have been negatively
testosterone-induced increases in muscle mass and correlated with waist/hip circumference ratio, visceral
strength can translate into improved physical func- fat area, glucose, insulin and C-peptide concentrations
tion and health outcomes in older men with mobility (Seidell et al. 1990; Khaw and Barrett-Connor 1992).
limitation or sarcopenia. The chapter will also
describe the mechanisms by which androgens
increase skeletal muscle mass, and strategies that
9.3 The effects of experimental
can be used to enhance the selectivity of androgen lowering of endogenous testosterone
action to improve the risk-to-benefit ratio. concentrations on body composition
The suppression of endogenous testosterone levels in
9.2 The association of circulating men by administration of a long-acting GnRH agon-
ist analog is associated with loss of fat-free mass, an
testosterone concentrations and muscle increase in fat mass, and a decrease in fractional
mass, muscle performance and physical muscle protein synthesis (Mauras et al. 1998). Testos-
function: epidemiological evidence terone suppression is also associated with a decrease
in whole body leucine oxidation as well as non-oxida-
Prepubertal boys and girls have similar muscle mass;
tive leucine disappearance rates (Mauras et al. 1998).
however, during the pubertal transition, the boys accrue
on average 4.5 kg greater muscle mass than girls, largely
due to the interactive effects of testosterone and GH. 9.4 The effects of physiological
Healthy, hypogonadal men have lower fat-free mass testosterone replacement in models
(Katznelson et al. 1996; 1998) and higher fat mass than
age-matched eugonadal men. Circulating testosterone of androgen deficiency
levels have been associated with appendicular muscle In all models of androgen deficiency that have been
mass and the strength of upper and lower extremity examined, testosterone replacement has been shown
muscles in men (Morley et al. 1997; Baumgartner et al. consistently to exert anabolic effects. For example, in
1998; Melton et al. 2000a; 2000b; Baumgartner pioneering studies conducted by Kochakian, testos-
2000; Roy et al. 2002). In epidemiological studies of terone replacement increased nitrogen retention in
community-dwelling older men, lower testosterone con- castrated males of several mammalian species
centrations are associated with reduced physical func- (Kochakian and Murlin 1935; Kochakian 1937;
tion, assessed using performance-based as well as 1950). Other studies reported nitrogen retention in
self-reported measures of physical function (van den eunuchoidal men, boys before puberty, and in women

192
Chapter 9: Androgen effects on the skeletal muscle

after testosterone supplementation (Kenyon et al. volume, does not change during testosterone admin-
1940). Several recent studies have re-examined the istration (Storer et al. 2003).
effects of testosterone on body composition and Plasma clearance rates of testosterone are lower in
muscle mass in hypogonadal men in more detail older men than in young men; consequently the
(Brodsky et al. 1996; Wang et al. 1996a; 2000; Bhasin increments in serum testosterone concentrations at
et al. 1997; Snyder et al. 2000; Steidle et al. 2003). any given testosterone dose are higher in older men
These studies are in agreement that replacement doses than in young men (Coviello et al. 2006). Older men
of testosterone, when administered to healthy, andro- also experience greater increments in hemoglobin and
gen-deficient men, increase fat-free mass, muscle size, hematocrit in response to testosterone administra-
and maximal voluntary strength (Brodsky et al. 1996; tion, even after adjusting for the higher testosterone
Wang et al. 1996a; 2000; Bhasin et al. 1997; Snyder levels (Calof et al. 2005; Coviello et al. 2008; Fernndez-
et al. 2000; Steidle et al. 2003). The muscle accretion Balsells et al. 2010). These age-related differences
during testosterone treatment has been reported to in hematocrit response to testosterone cannot be
increase in fractional muscle protein synthesis in explained easily on the basis of changes in erythropoi-
some studies (Brodsky et al. 1996; Mauras et al. 1998). etin or transferring receptor levels (Coviello et al. 2008).
We have shown recently that testosterone suppresses
9.5 Testosterone dose-response hepcidin, an important regulator of iron availability for
hematopoiesis, suggesting that testosterone might
relationships in young and older men stimulate erythropoiesis by increasing iron availability
Testosterone administration increases skeletal muscle (Bachman et al. 2010).
mass in healthy men; these anabolic effects of testos-
terone are related to the dose administered and the
circulating testosterone concentrations (Bhasin et al. 9.6 Randomized controlled trials of
2001; 2005; Storer et al. 2003; Woodhouse et al. 2003; testosterone and other androgens in
2004). In a randomized, masked study of testosterone
dose-response relationships, healthy young and older men with chronic diseases
men were treated with a long-acting GnRH agonist to The course of many chronic illnesses, such as chronic
suppress endogenous testosterone production and obstructive lung disease, end-stage renal disease, con-
given concurrently with one of several graded doses gestive heart failure, many types of cancers,
of testosterone enanthate, ranging from 25 mg intra- tuberculosis and HIV infection, are associated with
muscularly weekly to 600 mg intramuscularly weekly loss of lean body mass and strength, and increased
(Bhasin et al. 2001; 2005; Storer et al. 2003; Wood- risk of functional limitations, disability and poor dis-
house et al. 2003; 2004). Testosterone administration ease outcomes (MacAdams et al. 1986; Reid 1987;
was associated with dose-dependent increments in Coodley et al. 1994; Dobs et al. 1996; Grinspoon
fat-free mass, appendicular muscle mass and maximal et al. 1996; 2000; Arver et al. 1999; Salehian et al.
voluntary strength in the leg press exercise in healthy 1999; Rietschel et al. 2000; Bhasin et al. 2006).
young as well as older men (Bhasin et al. 2001; 2005; Accordingly, there has been considerable interest in
Storer et al. 2003; Woodhouse et al. 2003; 2004). In the application of androgens as anabolic therapy to
multivariate models, the gains in lean body mass and improve physical function and exercise capacity.
muscle size during testosterone administration could Several randomized trials have examined the effects
largely be explained by testosterone dose and the of androgen supplementation in men with chronic
circulating testosterone concentrations (Woodhouse illness (Buchwald et al. 1977; Berns et al. 1992; Schols
et al. 2003). Changes in whole-body, appendicular, et al. 1995; Reid et al. 1996; Coodley and Coodley 1997;
truncal, and intermuscular fat mass were inversely Grinspoon et al. 1998; Bhasin et al. 1998; 2000; Dobs
associated with testosterone dose and circulating tes- et al. 1999; Johansen 1999; Johansen et al. 1999; Painter
tosterone concentrations (Woodhouse et al. 2004). and Johansen 1999; Sattler et al. 1999; Strawford et al.
Unlike resistance exercise training, testosterone 1999a; 1999b; Crawford et al. 2003; Casaburi et al.
administration does not improve the contractile 2004). Meta-analyses of placebo-controlled random-
properties of skeletal muscle; thus specific force, that ized trials of testosterone replacement in HIV-infected
is, the maximal voluntary strength per unit muscle men with weight loss are in agreement that men

193
Chapter 9: Androgen effects on the skeletal muscle

assigned to the testosterone arm of these trials experi- 9.7 Randomizedtrials oftestosterone
enced greater gains in body weight and fat-free mass
than those randomized to the placebo arm (Kong and in healthy older men
Edmonds 2002; Johns et al. 2005; Bhasin et al. 2006). Meta-analyses of randomized clinical trials in commu-
However, the trials included in these meta-analyses nity dwelling, healthy, middle-aged and older men
were characterized by heterogeneous patient popula- have confirmed that testosterone therapy is associated
tions, small sample sizes and differences in testosterone with greater increments in fat-free mass and grip
dose and regimens. The three studies that showed gains strength, and greater reductions in fat mass than those
in fat-free mass selected patients with low testosterone associated with placebo administration alone (Fig. 9.1)
levels (Grinspoon et al. 1998; Bhasin et al. 1998; 2000). (Tenover 1992; Morley et al. 1993; Urban et al. 1995; Sih
In a placebo-controlled, randomized trial (Bhasin et al. et al. 1997; Ferrando et al. 1998; 2002; Clague et al. 1999;
2000), we demonstrated that testosterone replacement Snyder et al. 1999a; Tenover 1999; 2000; Kenny et al.
in HIV-infected men with low testosterone levels is 2001; Blackman et al. 2002; Johns et al. 2005; Page et al.
associated with significant gains in fat-free mass and 2005; Bhasin et al. 2006; Nair et al. 2006; Emmelot-
maximal voluntary strength in the leg press, leg curls, Vonk et al. 2008; LeBrasseur et al. 2009; Basaria et al.
bench press and latissimus dorsi pull-downs. Thus, 2010; Travison et al. 2011). Testosterone therapy
when the confounding influence of the learning effect improves self-reported physical function, such as that
is minimized and appropriate androgen-responsive measured using the physical function domain of the
measures of muscle strength are selected, testosterone MOS SF-36 (Snyder et al. 1999a). However, testoster-
replacement is associated with demonstrable increase one administration has not been shown to improve
in maximal voluntary strength in HIV-infected men performance-based measures of physical function in
with low testosterone levels. We do not know whether healthy older men, in spite of remarkable gains in
androgen replacement improves well-being, physical muscle mass and strength (Storer et al. 2008). The first
function, or other health-related outcomes in HIV- generation studies of testosterone were characterized
infected men. generally by their small sample size, inclusion of
The patients with end-stage renal disease who are healthy men without any functional limitations, and
on maintenance hemodialysis experience loss of skel- varying testosterone doses and regimens. The meas-
etal muscle mass, and decreased physical function and ures of physical function used in many trials had a low
exercise capacity (Johansen 1999; Johansen et al. ceiling (Bhasin et al. 2006).
1999; Painter and Johansen 1999). Nandrolone The doses of testosterone used in some of the
decanoate has been reported to increase hemoglobin trials were relatively low and were associated with
levels and fat-free mass in men with end-stage renal either small or no significant increments in testoster-
disease (Buchwald et al. 1977; Berns et al. 1992; one levels (Bhasin et al. 2006). As testosterone effects
Johansen et al. 1999). Testosterone replacement is on the skeletal muscle are related to testosterone dose
associated with a greater increase in fat-free mass, and circulating concentrations, it is possible that these
bone density, muscle strength, and quality of life than doses were insufficient to produce clinically meaning-
placebo in men receiving glucocorticoids (Reid et al. ful changes in muscle mass and strength.
1996; Crawford et al. 2003). Schols et al. (1995) have A major reason for the failure to demonstrate
reported modest increases in lean body mass and improvements in physical function is that testoster-
respiratory muscle strength with a low dose of nan- one trials of the first generation were conducted in
drolone in men and women with chronic obstructive healthy older men and used measures of physical
pulmonary disease (COPD). In a placebo-controlled, function that had a relatively low ceiling (Bhasin
randomized trial, Casaburi et al. (2004) demonstrated et al. 2006). The widely used measures such as
that testosterone therapy in men with COPD who 0.625 m stair climb, standing up from a chair, and
have low testosterone levels increases fat-free mass, 20 m walk are tasks that require only a small fraction
muscle size and maximal muscle strength to a greater of an individuals maximal voluntary strength. In most
extent than placebo. The effects of the combined healthy, older men, the baseline maximal voluntary
testosterone and resistance exercise training on strength is far higher than the threshold below which
muscle strength were greater than those of testoster- these measures would detect impairment. Given the
one alone (Casaburi et al. 2004). low intensity of the tasks used, it is not surprising that

194
Chapter 9: Androgen effects on the skeletal muscle

Kenny, 2001 performance on these tasks during testosterone


Symbol
administration. It also is possible that translation of
Combined
Harman, 2003
Individual
muscle mass and strength gains into functional
improvements may require functional training or
Wittert, 2003 cognitive and behavioral intervention. We do not
know whether task-specific training is necessary to
Snyder, 1999 induce the types of neuromuscular adaptations that
are necessary for improvements in physical function.
Tenover, 1992 The subjects recruited in the first generation trials of
testosterone were healthy community-dwelling men
Page, 2005 who did not have any functional limitations.
Several recent testosterone trials aimed at deter-
Ferrando, 2003 mining whether testosterone therapy improves phys-
ical function in older men with mobility limitations
Combined or one or more frailty characteristics (Kenny et al.
2001; Basaria et al. 2010; Srinivas-Shankar et al. 2010;
Travison et al. 2011). In a placebo-controlled trial
8.0 4.0 0.0 4.0 8.0 (Srinivas-Shankar et al. 2010), prefrail or frail older
men were randomized to either 50 mg testosterone gel
Mean difference in lean body mass change (kg)
or placebo gel for six months. The men randomized
to the testosterone arm experienced greater improve-
ments in isometric knee extension peak torque, lean
Wittert, 2003 body mass, and somatic and sexual symptoms than
Symbol
Combined those randomized to the placebo arm. Although
Individual measures of physical function did not differ signifi-
Sih, 1997
cantly between the two arms, they improved in the
subgroup of older and frail men (Srinivas-Shankar
Morley, 1993 et al. 2010). In a testosterone trial in older men with
mobility limitation (the TOM Trial; Basaria et al.
2010;Travison et al. 2011), the subjects were random-
Page, 2005 ized to either placebo or 10 g testosterone gel daily for
six months. The testosterone dose was adjusted to
achieve testosterone levels between 500 and 1000 ng/dl.
Combined
The men randomized to the testosterone arm experi-
enced greater improvements in leg-press strength,
chest-press strength and power, and loaded stair-
13.0 6.5 0.0 6.5 13.0 climbing speed and power than those assigned to
the placebo arm (Fig. 9.2). A greater proportion of
men in the testosterone arm improved more than the
Mean difference in grip strength change (kg)
minimal clinically important difference for leg-press
Fig. 9.1 A meta-analysis of the randomized trials of testosterone in and chest-press strength and stair-climbing speed
middle-aged and older men. The names of the first authors are than did in the placebo arm. The men in the testos-
listed. Overall, the men assigned to testosterone arms of the trials terone arm also experienced a higher frequency of
had greater gains in lean body mass than those assigned to the
placebo arms of the trial. Reproduced with permission from Bhasin adverse events and cardiovascular-related events than
et al. (2006). those in the placebo arm, leading the trials Data and
Safety Monitoring Board to recommend enrollment
relatively healthy older individuals recruited in these cessation and no further administration of study
initial testosterone trials showed neither impairment medication (Basaria et al. 2010; Travison et al.
in these threshold-dependent measures of physical 2011). Clinical trial data on the effects of testosterone
function at baseline, nor an improvement in on cardiovascular events are limited. The participants

195
Chapter 9: Androgen effects on the skeletal muscle

Testosterone preferred analyses are limited by the small size of most trials,
Leg press strength heterogeneity of subject populations, poor quality of
Chest press strength
adverse event reporting, and short treatment duration
in many trials. Most trials recruited healthy older men.
Leg press power Thus, the randomized trials are in agreement that
Chest press power testosterone increases muscle mass and strength.
However, substantial increases in lean body mass
Total LM
and leg-press strength during testosterone adminis-
ASLT tration have not been associated with consistent
Grip strength improvements in measures of physical function, such
as walking speed (Bhasin et al. 2006; Storer et al.
Unloaded gait speed
2008). Additionally, the findings of the TOM Trial
Loaded gait speed have raised concern that frail older persons with a
Unloaded stair climb high burden of chronic disease may be susceptible to
increased risk of adverse events during testosterone
Loaded stair climb
administration, especially at high testosterone con-
Lift and lower centrations (Basaria et al. 2010; Travison et al.
2011). Therefore, the therapeutic application of tes-
0.2 0.0 0.2 0.4 0.6 tosterone as a function-promoting anabolic therapy is
Treatment effect, SD units predicated crucially upon the development of strat-
egies which facilitate the translation of muscle mass
Fig. 9.2 The effects of testosterone on measures of muscle mass,
muscle performance and physical function in the Testosterone in and strength gains into functional improvements at
Older Men with Mobility Limitations Trial (the TOM Trial). Absolute lower testosterone concentrations that can be admin-
treatment differences (testosterone vs. placebo arms) are plotted for istered safely. Such adjunctive strategies might
the primary and secondary outcomes in units normalized to the
baseline standard deviation (SD) of measurement; point estimates include physical exercise interventions that emphasize
(red) are accompanied by 95% confidence intervals. Abbreviations: cognitive and behavioral training, or combined
ASLT, appendicular skeletal lean tissue mass; LM, lean mass. administration of testosterone with other anabolic
Reproduced with permission from Travison et al. 2011.
agents such as recombinant human growth hormone.
Empiric experience in athletes suggests that task-spe-
cific training is necessary for translating the muscle
in the TOM trial were older men (mean age 74) with mass and strength gains induced by androgen admin-
high prevalence of chronic conditions, such as heart istration into improvements in performance. Further
disease, diabetes, obesity, hypertension, and hyperlip- studies are needed to determine whether physical
idemia (Basaria et al. 2010; Travison et al. 2011). The activity interventions can facilitate the translation of
men assigned to the testosterone arm experienced testosterone-induced increases in muscle mass and
significantly higher frequencies of total adverse events, strength into clinically meaningful gains in physical
and cardiac, respiratory, and dermatological events function and health-related outcomes at lower testos-
than those assigned to the placebo group; these differ- terone doses that can be administered safely.
ences persisted after adjustment for baseline risk
factors (Basaria et al. 2010; Travison et al. 2011).
Men 75 years of age or older and men with higher 9.8 Testosterones effects on mood,
on-treatment testosterone levels appeared to be at fatigue/energy, sense of well-being and
greater risk of cardiovascular events. Meta-analyses
of randomized testosterone trials have reported quality of life
numerically greater number of cardiovascular events Epidemiological studies have reported an inconsistent
in the testosterone arms than in the placebo arms of association of testosterone levels with mood indices
trials, but these differences have not been statistically (Barrett-Connor et al. 1999; Margolese 2000; Seidman
significant presumably because of the small number of et al. 2001; 2002). Low bioavailable testosterone has
subjects enrolled in these trials (Calof et al. 2005; been weakly associated with depression in men (Barrett-
Fernndez-Balsells et al. 2010). However, these meta- Connor et al. 1999; Margolese 2000; Seidman

196
Chapter 9: Androgen effects on the skeletal muscle

et al. 2002). Testosterone appears to be associated more administration of higher doses is limited by the
with dysthymic mood than with major depressive dis- potential for adverse events, especially in older men
order (Seidman et al. 2002). Testosterone trials in men with high burdens of chronic conditions. Therefore,
with refractory depression taking antidepressants have strategies to achieve greater selectivity of action are
revealed conflicting results (Seidman and Rabkin 1998; necessary to realize the beneficial anabolic effects
Pope et al. 2003). without the undesirable adverse effects. Second,
In open-label trials, androgen administration in remarkable gains in skeletal muscle mass and strength
androgen-deficient men has been reported to improve induced by testosterone administration have not been
positive aspects of mood and reduce negative aspects associated consistently with improvements in physical
of mood (Wang et al. 1996b). Similar improvements function measures. Therefore, adjunctive strategies,
in mood and sense of well-being have been reported such as physical activity interventions, or cognitive
with testosterone replacement in surgically meno- and behavioral training, might be needed to induce
pausal women and women with adrenal insufficiency neuromuscular and behavioral adaptations that are
(Arlt et al. 1999; 2000; Shifren et al. 2000). necessary for translating muscle strength gains into
In HIV-infected men with low testosterone levels, clinically meaningful improvements in physical
testosterone supplementation has been reported to function.
restore libido and energy, and alleviate depressed Historically, three approaches have been used to
mood (Rabkin et al. 1995; 2000a; 2000b). There is achieve selectivity of hormone action: the elucidation
additional evidence that androgen improves energy of molecular targets in the signaling cascade of hor-
and reduces sense of fatigue in HIV-infected men mone action that might provide greater tissue select-
(Wagner et al. 1998; Knapp et al. 2008). ivity, the development of selective hormone receptor
The effects of short-term testosterone therapy on modulators, and the tissue-specific delivery of hor-
overall quality-of-life (QoL) scores using generic QoL mone. We will focus on the mechanism-based discov-
questionnaires have been inconsistent (Morley 2000; ery of more selective function-promoting anabolic
Reddy et al. 2000; Novak et al. 2002; Kenny et al. therapies.
2002). However, these studies did not have sufficient
power. Also, the QoL questionnaires used (e.g. SF-36) 9.10.1 Mechanisms by which testosterone
in previous studies are multidimensional and include
several domains that are not androgen responsive. increases skeletal muscle mass
Testosterone administration has been reported to There is general agreement that testosterones ana-
improve self-reported physical function (Snyder bolic effects on the skeletal muscle are mediated
et al. 1999a). through the AR (Fig. 9.3); the evidence for non-geno-
mic effects is weak. Testosterone-induced increase in
muscle mass is associated with hypertrophy of both
9.9 Reaction time type I and type II skeletal muscle fibers (Sinha-Hikim
Testosterone improves neuromuscular transmission et al. 2002). However, testosterone does not affect the
and reaction time in a frog hind leg model (Leslie absolute number or the relative proportion of type
et al. 1991; Blanco et al. 1997). It is possible that I and II muscle fibers (Sinha-Hikim et al. 2002).
testosterone may influence fall propensity by reducing Testosterone administration also is associated with a
reaction time. dose-related increase in the number of satellite cells
and myonuclei (Sinha-Hikim et al. 2003; 2006). How-
ever, the myonuclear domain the ratio of muscle
9.10 Strategies for achieving fiber cross-sectional area to myonuclear number
selectivity of testosterone action: does not change during testosterone administration.
dissociating the beneficial effects of Several hypotheses have been proposed to explain
the anabolic effects of testosterone on the skeletal
testosterone from its adverse effects muscle, and they are not mutually exclusive: stimula-
First, it is apparent that very substantial gains in tion of muscle protein synthesis, stimulation of the
skeletal muscle mass and strength are achievable with GH/IGF-1 axis, and the regulation of mesenchymal
supraphysiological doses of testosterone, and that the muscle progenitor cell differentiation.

197
Chapter 9: Androgen effects on the skeletal muscle

Multipotent
progenitor cells

Satellite cells

Differentiated
myocyte

OH
AR im IGF-1
CH CH3
CH3
CH3

SRD5A O
O H IGF-1R
DHT
T Muscle
protein
Atrogens/FoXo synthesis
Ubiquitin/
proteasome

GH IGF-1
Fig. 9.3 Potential mechanisms of the anabolic effects of testosterone on the skeletal muscle. Testosterone (T) could potentially increase
muscle mass through multiple mechanisms. The vast majority of evidence suggests that testosterones effects are mediated through the AR.
It does not appear that 5-alpha reduction of testosterone is essential for mediating testosterones effects on the muscle. Testosterone
stimulates GH secretion and increases IGF-1 levels, but circulating GH and IGF-1 are not essential for mediating testosterones effects on the
muscle. Testosterone promotes the differentiation of mesenchymal multipotent progenitor cell into the myogenic lineage. Whether
testosterone has direct effects on muscle protein synthesis, muscle protein degradation or atrophy pathways remains to be investigated.
Testosterones effect on proliferation and differentiation of skeletal muscle progenitor cells requires the mediation of IGF-1R signaling. See plate
section for color version.

The observation that testosterone and other the skeletal muscle, predominantly in the satellite
androgens increase nitrogen retention in androgen- cells, identified by their location outside the sarco-
deficient men (Kochakian and Murlin 1935; Kocha- lemma but inside the lamina, and by c-MET, and
kian 1937; 1950; Kenyon et al. 1940) led to the CD34 staining (Sinha-Hikim et al. 2004). Androgen
hypothesis that testosterone stimulates muscle protein receptor protein expression is also observed in many
synthesis. Several investigators using stable isotopes myonuclei, and in CD34+ cells outside the lamina,
have shown that testosterone therapy improves frac- vascular endothelial cells and myofibroblasts (Sinha-
tional muscle protein synthesis and reutilization of Hikim et al. 2004).
amino acids (Brodsky et al. 1996; Mauras et al. 1998; We determined the effects of testosterone and
Sheffield-Moore et al. 1999; 2006; Bhasin et al. 2003). DHT on the differentiation of multipotent,
The effects of testosterone on muscle protein degrad- mesenchymal C3H10T1/2 cells (Singh et al. 2003;
ation are less clear. 2009). Although untreated cells express low levels
The muscle protein synthesis hypothesis does not of AR protein, DHT and testosterone upregulate
easily explain the reciprocal change in fat mass or the AR expression in these cells. Incubation with testos-
increased number of satellite cells in testosterone- terone and DHT increases the number of MyoD+
treated men (Sinha-Hikim et al. 2003; 2004; 2006). myogenic cells and MHC+ myotubes (Singh et al.
Therefore, we considered the alternate hypothesis that 2003; 2009). Testosterone and DHT upregulate the
testosterone might regulate the differentiation of expression of MyoD, and MHC mRNA and protein
mesenchymal multipotent cells, promoting their dif- levels increased dose dependently. Both testosterone
ferentiation into the myogenic lineage and inhibiting and DHT also decrease the number of oil red
adipogenic differentiation. Androgen receptor pro- O positive adipocytes and downregulate the expres-
tein is expressed in mesenchymal progenitor cells in sion of PPARg2 mRNA and PPARg2 and C/EBPa

198
Chapter 9: Androgen effects on the skeletal muscle

proteins that are markers of adipogenic differenti- myogenesis (Singh et al. 2003; 2006; 2009). The
ation (Singh et al. 2003; 2009; Gupta et al. 2009). The androgen signal is cross-communicated to the TGF-
effects of testosterone and DHT on myogenesis and b/SMAD pathway through follistatin (Singh et al.
adipogenesis are blocked by bicalutamide, an andro- 2009), which blocks TGF-b/SMAD signaling in vivo
gen receptor antagonist (Singh et al. 2003; 2009). and in vitro. When administered to castrated mice,
Hence, testosterone and DHT regulate the differenti- follistatin increases skeletal muscle mass and
ation of mesenchymal multipotent cells by promot- decreases fat mass without affecting the prostate.
ing their differentiation into the myogenic lineage Thus, follistatin, a mediator of androgen action, may
and inhibiting their differentiation into adipocytes have potential as a selective function-promoting ana-
through an AR-mediated pathway (Singh et al. bolic therapy that might increase skeletal muscle mass
2003; 2009). without affecting the prostate.
In separate studies, we have shown that DHT In a myocyte-specific AR knock-out (mARKO)
also regulates the differentiation of human marrow- mouse, lean body mass is lower in the mARKO than
derived, mesenchymal stem cells (hMSCs) from in control mice (Ophoff et al. 2009). The weight of the
adult men (Gupta et al. 2009). Dihydrotestosterone androgen-sensitive levator ani is significantly reduced
upregulates AR expression and inhibits lipid (46%); whereas the weights of other muscle groups
accumulation in adipocytes differentiated from were slightly reduced (Reddy et al. 2000), suggesting
hMSCs and downregulates the expression of aP2, that in addition to its effects on muscle progenitor
PPARg, leptin and C/EBPa (Gupta et al. 2009). cells, testosterone affects post-mitotic myocytes.
Bicalutamide attenuates DHTs inhibitory effects
on adipogenic differentiation of hMSCs. Adipocytes
differentiated in the presence of DHT accumulate
9.10.2 The role of circulating growth
smaller oil droplets, suggesting reduced extent of hormone and IGF-1, and intramuscular IGF-1
maturation. Dihydrotestosterone decreases the signaling in mediating testosterones effects
incorporation of labeled fatty acid into triglyceride,
and downregulates acetyl CoA carboxylase and on the muscle
DGAT2 expression in adipocytes derived from Testosterone stimulates pulsatile GH secretion and
hMSCs. Thus, DHT inhibits adipogenic differenti- increases serum IGF-1 concentrations in peripuber-
ation of hMSCs through an AR-mediated pathway tal boys and in boys with constitutional delay of
(Gupta et al. 2009)). puberty (Eakman et al. 1996; Giustina et al. 1997;
WNT signaling plays an important role in regu- Bondanelli et al. 2003). Androgen administration
lating the differentiation of mesenchymal progeni- has also been shown to increase circulating IGF-1
tor cells, and testosterone and DHT promote the levels and upregulate intramuscular IGF-1 mRNA
association of liganded androgen receptor with expression in men (Ferrando et al. 2002). However,
b-catenin, stabilizing the latter and causing the in castrated, hypophysectomized mice, testosterone
androgen receptorb-catenin complex to translo- increases muscle mass, indicating that circulating
cate into the nucleus and activate a number of GH and IGF-1 are not essential for mediating
WNT target genes (Singh et al. 2006; 2009). Double the anabolic effects of testosterone on the muscle
immunofluorescence and immunoprecipitation (Serra et al. 2011).
studies have revealed that AR, b-catenin, and Testosterone stimulates the proliferation of
TCF-4 are colocalized in the nucleus in both testos- human skeletal muscle cells in vitro: an effect blocked
terone-treated (100 nM) and DHT-treated (10 nM) by small interference RNA targeting human IGF-1
cells (Singh et al. 2006; 2009), suggesting that they receptor (IGF-1R) (sihIGF-1R) (Serra et al. 2011). In
interact to form a complex. Both b-catenin and differentiation conditions, testosterone promotes the
TCF-4 play an essential role in mediating androgen fusion of human skeletal muscle cells into larger
effects on the differentiation of C3H10T1/2 cells myotubes, an effect attenuated by sihIGF-1R (Serra
(Singh et al. 2009). et al. 2011). Thus, intramuscular IGF-1 signaling
Testosterone regulates the expression of several plays an important role in mediating testosterones
WNT target genes, including follistatin, which plays effects on skeletal muscle progenitor cell growth and
an essential role in mediating testosterones effects on differentiation (Serra et al. 2011)).

199
Chapter 9: Androgen effects on the skeletal muscle

9.11 Perspective on the future of  Testosterone acts through multiple


testosterone as a function-promoting mechanisms to increase skeletal muscle mass;
it promotes the differentiation of
therapy mesenchymal multipotent cells into myogenic
Strong clinical trial data support the view that andro- lineage and inhibits their differentiation into
gens increase skeletal muscle mass and maximal vol- adipogenic lineage through an AR-mediated
untary strength. However, we do not know whether pathway that involves cross-communication
testosterone administration improves physical func- between the WNT-b-catenin and TGF-b
tion and health outcomes and reduces disability in signaling pathways. Testosterone stimulates
GH and IGF-1 secretion, but circulating GH and
older individuals with functional limitations. Add-
IGF-1 are not essential for mediating the
itionally, there are unresolved concerns about the anabolic effects of testosterone on the
potential long-term adverse effects of testosterone on muscle. Intramuscular IGF-1 receptor
cardiovascular events and the prostate. Strategies to signaling plays an important role in mediating
increase the selectivity of androgen action are needed the anabolic effects of testosterone on the
to achieve a higher risk-to-benefit ratio. Additionally, muscle.
it is possible that risk-factor-based multimodality  In spite of unequivocal increases in muscle
interventions that include physical activity interven- mass and strength, testosterone has not been
tions or combination of anabolic therapies might be shown to improve performance-based measures
needed to improve physical function at testosterone of physical function and health outcomes.
doses that can be safely administered. Given the uncer- Physical activity interventions that
incorporate cognitive and behavioral therapy,
tainty about the regulatory approval pathway and con-
or multimodality interventions that include
cerns about long-term safety, the initial efficacy trials one or more anabolic therapies based on
are likely to be conducted in the treatment mode rather risk-factor identification might be needed to
than in the prevention mode and for short-term indi- translate testosterone-induced muscle mass
cations in conditions associated with grievous out- and strength gains into functional
comes, such as cancer cachexia and hip fracture, and improvements.
to promote rehabilitation after an acute illness.  Neither the clinical benefits nor the long-term
risks of testosterone therapy are known.
Frail elderly most in need of anabolic therapy
9.12 Key messages
may also be at increased risk of cardiovascular
 There is an enormous unmet need for adverse events. Strategies to dissociate the
function-promoting anabolic therapies for beneficial anabolic effects of testosterone
functional limitations associated with aging and from its adverse effects are needed. Such
acute and chronic illnesses. strategies might include the development of
 Randomized trials are in agreement that testoster- selective androgen receptor modulators,
one administration increases skeletal muscle mass mechanism-specific therapies and tissue-specific
and maximal voluntary strength; these effects are delivery.
related to testosterone dose and concentrations.

9.13 References adrenal insufficiency Bachman E, Feng R, Travison T, Li M,


pharmacokinetics, bioconversion Olbina G, Ostland V, Ulloor J,
Arlt W, Callies F, van Vlijmen JC, and clinical effects on well-being, Zhang A, Basaria S, Ganz T,
Koehler I, Reincke M, sexuality and cognition. Endocr Res Westerman M, Bhasin S (2010)
Bidlingmaier M, Huebler D, 26:505511 Testosterone suppresses hepcidin in
Oettel M, Ernst M, Schulte HM, men: a potential mechanism for
Allolio B (1999) Arver S, Sinha-Hikim I, Beall G, testosterone-induced
Dehydroepiandrosterone Guerrero M, Shen R, Bhasin S erythrocytosis. J Clin Endocrinol
replacement in women with (1999) Serum dihydrotestosterone Metab 95:47434747
adrenal insufficiency. N Engl J Med and testosterone concentrations
341:10131020 in human immunodeficiency virus- Barrett-Connor E, von Muhlen DG,
infected men with and without Kritz-Silverstein D (1999)
Arlt W, Callies F, Allolio B (2000) Bioavailable testosterone and
DHEA replacement in women with weight loss. J Androl 20:611618

200
Chapter 9: Androgen effects on the skeletal muscle

depressed mood in older men: Effects of testosterone replacement Stevens TE, Jayme J, OConnor KG,
the Rancho Bernardo Study. with a nongenital, ransdermal Christmas C, Tobin JD, Stewart KJ,
J Clin Endocrinol Metab system, Androderm, in human Cottrell E, St Clair C, Pabst KM,
84:573577 immunodeficiency virus-infected Harman SM (2002) Growth
Basaria S, Coviello AD, Travison TG, men with low testosterone levels. hormone and sex steroid
Storer TW, Farwell WR, Jette AM, J Clin Endocrinol Metab 83: administration in healthy aged
Eder R, Tennstedt S, Ulloor J, 31553162 women and men: a randomized
Zhang A, Choong K, Lakshman Bhasin S, Storer TW, Javanbakht M, controlled trial. JAMA 288:
KM, Mazer NA, Miciek R, Krasnoff Berman N, Yarasheski KE, Phillips 22822292
J, Elmi A, Knapp PE, Brooks B, J, Dike M, Sinha-Hikim I, Shen R, Blanco CE, Popper P, Micevych P
Appleman E, Aggarwal S, Bhasin G, Hays RD, Beall G (2000) (1997) Anabolic-androgenic steroid
Hede-Brierley L, Bhatia A, Testosterone replacement and induced alterations in choline
Collins L, LeBrasseur N, Fiore LD, resistance exercise in HIV-infected acetyltransferase messenger RNA
Bhasin S (2010) Adverse events men with weight loss and low levels of spinal cord motoneurons
associated with testosterone testosterone levels. JAMA 283: in the male rat. Neuroscience
administration. N Engl J Med 763770 78:873882
363:109122 Bhasin S, Woodhouse L, Casaburi R, Bondanelli M, Ambrosio MR, Margutti
Baumgartner RN (2000) Body Singh AB, Bhasin D, Berman N, A, Franceschetti P, Zatelli MC, degli
composition in healthy Chen X, Yarasheski KE, Magliano L, Uberti EC (2003) Activation of the
aging. Ann N Y Acad Sci Dzekov C, Dzekov J, Bross R, somatotropic axis by testosterone in
904:437448 Phillips J, Sinha-Hikim I, Shen R, adult men: evidence for a role of
Storer TW (2001) Testosterone hypothalamic growth hormone-
Baumgartner RN, Koehler KM,
dose-response relationships in releasing hormone.
Gallagher D, Romero L, Heymsfield
healthy young men. Am J Physiol Neuroendocrinology 77:380387
SB, Ross RR, Garry PJ, Lindeman
Endocrinol Metab 281:E1172E1181
RD (1998) Epidemiology of Brodsky IG, Balagopal P, Nair KS
sarcopenia among the elderly in Bhasin S, Taylor WE, Singh R, Artaza J, (1996) Effects of testosterone
New Mexico. Am J Epidemiol Sinha-Hikim I, Jasuja R, Choi H, replacement on muscle mass and
147:755763 Gonzalez-Cadavid NF (2003) The muscle protein synthesis in
mechanisms of androgen effects hypogonadal men-a clinical
Berns JS, Rudnick MR, Cohen RM
on body composition: mesenchymal research center study. J Clin
(1992) A controlled trial of
pluripotent cell as the target of Endocrinol Metab 81:34693475
recombinant human erythropoietin
androgen action. J Gerontol
and nandrolone decanoate in the
A Biol Sci Med Sci 58: Buchwald D, Argyres S, Easterling RE,
treatment of anemia in patients on Oelshlegel FJ Jr, Brewer GJ,
M1103M1110
chronic hemodialysis. Clin Nephrol Schoomaker EB, Abbrecht PH,
37:264267 Bhasin S, Woodhouse L, Casaburi R,
Williams GW, Weller JM (1977)
Singh AB, Mac RP, Lee M,
Bhasin S, Storer TW, Berman N, Effect of nandrolone decanoate
Yarasheski KE, Sinha-Hikim I,
Callegari C, Clevenger B, Phillips J, on the anemia of chronic
Dzekov C, Dzekov J, Magliano L,
Bunnell TJ, Tricker R, Shirazi A, hemodialysis patients. Nephron
Storer TW (2005) Older men are as
Casaburi R (1996) The effects of 18:232238
responsive as young men to the
supraphysiologic doses of anabolic effects of graded doses Calof OM, Singh AB, Lee ML, Kenny
testosterone on muscle size and of testosterone on the skeletal AM, Urban RJ, Tenover JL, Bhasin
strength in normal men. N Engl muscle. J Clin Endocrinol Metab S (2005) Adverse events associated
J Med 335:17 90:678688 with testosterone replacement in
Bhasin S, Storer TW, Berman N, Bhasin S, Calof OM, Storer TW, Lee middle-aged and older men: a
Yarasheski KE, Clevenger B, Phillips ML, Mazer NA, Jasuja R, Montori meta-analysis of randomized,
J, Lee WP, Bunnell TJ, Casaburi R VM, Gao W, Dalton JT (2006) placebo-controlled trials.
(1997) Testosterone replacement Drug insight: testosterone and J Gerontol A Biol Sci Med Sci
increases fat-free mass and selective androgen receptor 60:14511457
muscle size in hypogonadal modulators as anabolic therapies Casaburi R, Bhasin S, Cosentino L,
men. J Clin Endocrinol Metab for chronic illness and aging. Porszasz J, Somfay A, Lewis MI,
82:407413 Nat Clin Pract Endocrinol Metab Fournier M, Storer TW (2004)
Bhasin S, Storer TW, Asbel-Sethi N, 2:146159 Effects of testosterone and
Kilbourne A, Hays R, Sinha-Hikim Blackman MR, Sorkin JD, Munzer T, resistance training in men with
I, Shen R, Arver S, Beall G (1998) Bellantoni MF, Busby-Whitehead J, chronic obstructive pulmonary

201
Chapter 9: Androgen effects on the skeletal muscle

disease. Am J Respir Crit Care Med wasting. J Clin Endocrinol Metab testosterone. J Urol 165:
170:870878 81:41084112 371373
Cawthon PM, Ensrud KE, Laughlin Dobs AS, Cofrancesco J, Nolten WE, Giustina A, Scalvini T, Tassi C,
GA, Cauley JA, Dam TT, Barrett- Danoff A, Anderson R, Hamilton Desenzani P, Poiesi C, Wehrenberg
Connor E, Fink HA, Hoffman AR, CD, Feinberg J, Seekins D, Yangco WB, Rogol AD, Veldhuis JD (1997)
Lau E, Lane NE, Stefanick ML, B, Rhame F (1999) The use of a Maturation of the regulation of
Cummings SR, Orwoll ES; transscrotal testosterone delivery growth hormone secretion in young
Osteoporotic Fractures in Men system in the treatment of patients males with hypogonadotropic
(MrOS) Research Group (2009) Sex with weight loss related to human hypogonadism pharmacologically
hormones and frailty in older men: immunodeficiency virus infection. exposed to progressive increments
the osteoporotic fractures in men Am J Med 107:126132 in serum testosterone. J Clin
(MrOS) study. J Clin Endocrinol Eakman GD, Dallas JS, Ponder SW, Endocrinol Metab 82:12101219
Metab 94:38063815 Keenan BS (1996) The effects of Grinspoon S, Corcoran C, Lee K,
Clague JE, Wu FC, Horan MA (1999) testosterone and Burrows B, Hubbard J, Katznelson
Difficulties in measuring the effect dihydrotestosterone on L, Walsh M, Guccione A, Cannan J,
of testosterone replacement therapy hypothalamic regulation of growth Heller H, Basgoz N, Klibanski A
on muscle function in older men. hormone secretion. J Clin (1996) Loss of lean body and muscle
Int J Androl 22:261265 Endocrinol Metab 81:12171223 mass correlates with androgen levels
Coodley GO, Coodley MK (1997) Emmelot-Vonk MH, Verhaar HJ, in hypogonadal men with acquired
A trial of testosterone therapy for Nakhai Pour HR, Aleman A, Lock immunodeficiency syndrome and
HIV-associated weight loss. Aids TM, Bosch JL, Grobbee DE, van der wasting. J Clin Endocrinol Metab
11:13471352 Schouw YT (2008) Effect of 81:40514058
Coodley GO, Loveless MO, Nelson testosterone supplementation on Grinspoon S, Corcoran C, Askari H,
HD, Coodley MK (1994) Endocrine functional mobility, cognition, and Schoenfeld D, Wolf L, Burrows B,
function in the HIV wasting other parameters in older men: a Walsh M, Hayden D, Parlman K,
syndrome. J Acquir Immune Defic randomized controlled trial. JAMA Anderson E, Basgoz N, Klibanski A
Syndr 7:4651 299:3952 (1998) Effects of androgen
Fernndez-Balsells MM, Murad MH, administration in men with the AIDS
Coviello AD, Lakshman K, Mazer NA,
Lane M, Lampropulos JF, wasting syndrome. A randomized,
Bhasin S (2006) Differences in the
Albuquerque F, Mullan RJ, Agrwal double-blind, placebo-controlled
apparent metabolic clearance rate of
N, Elamin MB, Gallegos-Orozco JF, trial. Ann Intern Med 129:1826
testosterone in young and older
men with gonadotropin suppression Wang AT, Erwin PJ, Bhasin S, Grinspoon S, Corcoran C, Stanley T,
receiving graded doses of Montori VM (2010) Clinical review Baaj A, Basgoz N, Klibanski A
testosterone. J Clin Endocrinol 1: adverse effects of testosterone (2000) Effects of hypogonadism and
Metab 91:46694675 therapy in adult men: a systematic testosterone administration on
review and meta-analysis. J Clin depression indices in HIV-infected
Coviello AD, Kaplan B, Lakshman KM,
Endocrinol Metab 95:25602575 men. J Clin Endocrinol Metab
Chen T, Singh AB, Bhasin S (2008)
Ferrando AA, Tipton KD, Doyle D, 85:6065
Effects of graded doses of
testosterone on erythropoiesis in Phillips SM, Cortiella J, Wolfe RR Gupta V, Bhasin S, Guo W, Singh R,
healthy young and older men. J Clin (1998) Testosterone injection Miki R, Chauhan P, Choong K,
Endocrinol Metab 93:914919 stimulates net protein synthesis but Tchkonia T, Lebrasseur NK,
Crawford BA, Liu PY, Kean MT, not tissue amino acid transport. Am Flanagan JN, Hamilton JA, Viereck
Bleasel JF, Handelsman DJ (2003) J Physiol 275:E864E871 JC, Narula NS, Kirkland JL, Jasuja R
Randomized placebo-controlled Ferrando AA, Sheffield-Moore M, (2009) Effects of dihydrotestosterone
trial of androgen effects on muscle Yeckel CW, Gilkison C, Jiang J, on differentiation and proliferation
and bone in men requiring long- Achacosa A, Lieberman SA, Tipton of human mesenchymal stem cells
term systemic glucocorticoid K, Wolfe RR, Urban RJ (2002) and preadipocytes. Mol Cell
treatment. J Clin Endocrinol Metab Testosterone administration to older Endocrinol 296:3240
88:31673176 men improves muscle function: Johansen KL (1999) Physical
Dobs AS, Few WL 3rd, Blackman MR, molecular and physiological functioning and exercise capacity in
Harman SM, Hoover DR, Graham mechanisms. Am J Physiol patients on dialysis. Adv Ren
NM (1996) Serum hormones in Endocrinol Metab 282:E601E607 Replace Ther 6:141148
men with human Freeman ER, Bloom DA, McGuire EJ Johansen KL, Mulligan K, Schambelan
immunodeficiency virus-associated (2001) A brief history of M (1999) Anabolic effects of

202
Chapter 9: Androgen effects on the skeletal muscle

nandrolone decanoate in patients of testosterone on physical function, maleis there a link? J Geriatr
receiving dialysis: a randomized muscle performance, mood, and Psychiatry Neurol 13:93101
controlled trial. Jama 281:12751281 fatigue in men with HIV-associated Mauras N, Hayes V, Welch S, Rini A,
Johns K, Beddall MJ, Corrin RC (2005) weight loss. Am J Physiol Endocrinol Helgeson K, Dokler M, Veldhuis JD,
Anabolic steroids for the treatment Metab 294:E1135E1143 Urban RJ (1998) Testosterone
of weight loss in HIV-infected Kochakian C (1937) Testosterone and deficiency in young men: marked
individuals. Cochrane Database Syst testosterone acetate and the protein alterations in whole body protein
Rev 19:CD005483 and energy metabolism of castrate kinetics, strength, and adiposity. J Clin
Katznelson L, Finkelstein JS, dogs. Endocrinology 21:750755 Endocrinol Metab 83:18861892
Schoenfeld DA, Rosenthal DI, Kochakian C (1950) Comparison of Melton LJ 3rd, Khosla S, Crowson CS,
Anderson EJ, Klibanski A (1996) protein anabolic property of various OConnor MK, OFallon WM,
Increase in bone density and lean androgens in the castrated rat. Am Riggs BL (2000a) Epidemiology of
body mass during testosterone J Physiol 60:5358 sarcopenia. J Am Geriatr Soc
administration in men with
Kochakian C, Murlin J (1935) The effect 48:625630
acquired hypogonadism. J Clin
of male hormone on the protein and Melton LJ 3rd, Khosla S, Riggs BL
Endocrinol Metab 81:43584365
energy metabolism of castrate dogs. (2000b) Epidemiology of sarcopenia.
Katznelson L, Rosenthal DI, Rosol MS, J Nutrition 10:437459 Mayo Clin Proc 75 (Suppl):S10S12;
Anderson EJ, Hayden DL, discussion:S12S13
Schoenfeld DA, Klibanski A (1998) Kong A, Edmonds P (2002)
Using quantitative CT to assess Testosterone therapy in HIV Morley JE (2000) Andropause,
adipose distribution in adult men wasting syndrome: systematic testosterone therapy, and quality of
with acquired hypogonadism. Am review and meta-analysis. Lancet life in aging men. Cleve Clin J Med
J Roentgenol 170:423427 Infect Dis 2:692699 67:880882
Kenny AM, Prestwood KM, Gruman Krasnoff J, Basaria S, Pencina M, Kaur Morley JE, Perry HM 3rd, Kaiser FE,
CA, Marcello KM, Raisz LG (2001) G, Coviello A, Vasan R, Bhasin S, Kraenzle D, Jensen J, Houston K,
Effects of transdermal testosterone Murabito JM (2009) Free Mattammal M, Perry HM Jr (1993)
on bone and muscle in older men testosterone levels are associated Effects of testosterone replacement
with low bioavailable testosterone with mobility limitation and therapy in old hypogonadal males: a
levels. J Gerontol A Biol Sci Med Sci physical performance in preliminary study. J Am Geriatr Soc
56:M266M272 community-dwelling men: the 41:149152
Framingham Offspring Study. J Clin Morley JE, Kaiser FE, Perry HM 3rd,
Kenny AM, Bellantonio S, Gruman Endocrinol Metab 95:27902799
CA, Acosta RD, Prestwood KM Patrick P, Morley PM, Stauber PM,
(2002) Effects of transdermal LeBrasseur NK, Lajevardi N, Miciek R, Vellas B, Baumgartner RN, Garry PJ
testosterone on cognitive function Mazer N, Storer TW, Bhasin S (1997) Longitudinal changes in
and health perception in older men (2009) Effects of testosterone testosterone, luteinizing hormone,
with low bioavailable testosterone therapy on muscle performance and and follicle-stimulating hormone in
levels. J Gerontol A Biol Sci Med Sci physical function in older men with healthy older men. Metabolism
57:M321M325 mobility limitations (The TOM 46:410413
Trial): design and methods. Nair KS, Rizza RA, OBrien P,
Kenyon A, Knowlton K, Sandiford I,
Contemp Clin Trials 30:133140 Dhatariya K, Short KR, Nehra A,
Koch FC, Lotwin G (1940)
A comparative study of the Leslie M, Forger NG, Breedlove SM Vittone JL, Klee GG, Basu A, Basu
metabolic effects of testosterone (1991) Sexual dimorphism and R, Cobelli C, Toffolo G, Dalla Man
propionate in normal men and androgen effects on spinal C, Tindall DJ, Melton LJ 3rd, Smith
women and in eunuchoidism. motoneurons innervating the rat GE, Khosla S, Jensen MD (2006)
Endocrinology 26:2645 flexor digitorum brevis. Brain Res DHEA in elderly women and
Khaw KT, Barrett-Connor E (1992) 561:269273 DHEA or testosterone in elderly
Lower endogenous androgens MacAdams MR, White RH, Chipps BE men. N Engl J Med 355:16471659
predict central adiposity in men. (1986) Reduction of serum Novak A, Brod M, Elbers J (2002)
Ann Epidemiol 2:675682 testosterone levels during chronic Andropause and quality of life:
Knapp PE, Storer TW, Herbst KL, glucocorticoid therapy. Ann Intern findings from patient focus groups
Singh AB, Dzekov C, Dzekov J, Med 104:648651 and clinical experts. Maturitas
LaValley M, Zhang A, Ulloor J, Margolese HC (2000) The male 43:231237
Bhasin S (2008) Effects of a menopause and mood: testosterone Ophoff J, Van Proeyen K, Callewaert F,
supraphysiological dose decline and depression in the aging De Gendt K, De Bock K, Vanden

203
Chapter 9: Androgen effects on the skeletal muscle

Bosch A, Verhoeven G, Hespel P, males a pilot study. J Clin Pharm support and anabolic steroids in
Vanderschueren D (2009) Androgen Ther 25:421426 patients with chronic obstructive
signaling in myocytes contributes to Reid IR (1987) Serum testosterone pulmonary disease. A placebo-
the maintenance of muscle mass and levels during chronic glucocorticoid controlled randomized trial.
fiber type regulation but not to therapy. Ann Intern Med Am J Respir Crit Care Med
muscle strength or fatigue. 106:639640 152:12681274
Endocrinology 150:35583566 Seidell JC, Bjorntorp P, Sjostrom L,
Reid IR, Wattie DJ, Evans MC,
Orwoll E, Lambert LC, Marshall LM, Stapleton JP (1996) Testosterone Kvist H, Sannerstedt R (1990)
Blank J, Barrett-Connor E, Cauley J, therapy in glucocorticoid-treated Visceral fat accumulation in men is
Ensrud K, Cummings SR (2006) men. Arch Intern Med positively associated with insulin,
Endogenous testosterone levels, 156:11731177 glucose, and C-peptide levels, but
physical performance, and fall risk negatively with testosterone levels.
in older men. Arch Intern Med Rietschel P, Corcoran C, Stanley T, Metabolism 39:897901
166:21242131 Basgoz N, Klibanski A, Grinspoon S
(2000) Prevalence of hypogonadism Seidman SN, Rabkin JG (1998)
Page ST, Amory JK, Bowman FD, among men with weight loss related Testosterone replacement therapy
Anawalt BD, Matsumoto AM, to human immunodeficiency virus for hypogonadal men with SSRI-
Bremner WJ, Tenover JL (2005) infection who were receiving highly refractory depression. J Affect
Exogenous testosterone (T) alone or active antiretroviral therapy. Clin Disord 48:157161
with finasteride increases physical Infect Dis 31:12401244 Seidman SN, Araujo AB, Roose SP,
performance, grip strength, and
Roy TA, Blackman MR, Harman SM, McKinlay JB (2001) Testosterone
lean body mass in older men with
Tobin JD, Schrager M, Metter EJ level, androgen receptor
low serum T. J Clin Endocrinol
(2002) Interrelationships of serum polymorphism, and depressive
Metab 90:15021510
testosterone and free testosterone symptoms in middle-aged men. Biol
Painter P, Johansen K (1999) Physical index with FFM and strength in Psychiatry 50:371376
functioning in end-stage renal aging men. Am J Physiol Endocrinol
disease. Introduction: a call to Seidman SN, Araujo AB, Roose SP,
Metab 283:E284E294 Devanand DP, Xie S, Cooper TB,
activity. Adv Ren Replace Ther
6:107109 Salehian B, Jacobson D, Swerdloff RS, McKinlay JB (2002) Low
Grafe MR, Sinha-Hikim I, testosterone levels in elderly men
Pope HG Jr, Cohane GH, Kanayama G, McCutchan JA (1999) Testicular with dysthymic disorder. Am
Siegel AJ, Hudson JI (2003) pathologic changes and the J Psychiatry 159:456459
Testosterone gel supplementation pituitary-testicular axis during
for men with refractory depression: Serra C, Bhasin S, Tangherlini F,
human immunodeficiency virus Barton ER, Ganno M, Zhang A,
a randomized, placebo-controlled infection. Endocr Pract 5:19
trial. Am J Psychiatry 160:105111 Shansky J, Vandenburgh HH,
Sattler FR, Jaque SV, Schroeder ET, Travison TG, Jasuja R, Morris C
Rabkin JG, Rabkin R, Wagner G (1995) Olson C, Dube MP, Martinez C, (2011) The role of GH and IGF-I in
Testosterone replacement therapy Briggs W, Horton R, Azen S (1999) mediating anabolic effects of
in HIV illness. Gen Hosp Psychiatry Effects of pharmacological doses of testosterone on androgen-
17:3742 nandrolone decanoate and responsive muscle. Endocrinology
Rabkin JG, Ferrando SJ, Wagner GJ, progressive resistance training in 152:193206
Rabkin R (2000a) DHEA treatment immunodeficient patients infected Sheffield-Moore M, Urban RJ, Wolf
for HIV+ patients: effects on mood, with human immunodeficiency SE, Jiang J, Catlin DH, Herndon
androgenic and anabolic parameters. virus. J Clin Endocrinol Metab DN, Wolfe RR, Ferrando AA (1999)
Psychoneuroendocrinology 25:5368 84:12681276 Short-term oxandrolone
Rabkin JG, Wagner GJ, Rabkin R Schaap LA, Pluijm SM, Smit JH, van administration stimulates net
(2000b) A double-blind, placebo- Schoor NM, Visser M, Gooren LJ, muscle protein synthesis in young
controlled trial of testosterone Lips P (2005) The association of sex men. J Clin Endocrinol Metab
therapy for HIV-positive men with hormone levels with poor mobility, 84:27052711
hypogonadal symptoms. Arch Gen low muscle strength and incidence Sheffield-Moore M, Paddon-Jones D,
Psychiatry 57:141147; of falls among older men and Casperson SL, Gilkison C, Volpi E,
discussion:155146 women. Clin Endocrinol (Oxf) Wolf SE, Jiang J, Rosenblatt JI,
Reddy P, White CM, Dunn AB, Moyna 63:152160 Urban RJ (2006) Androgen therapy
NM, Thompson PD (2000) The Schols AM, Soeters PB, Mostert R, induces muscle protein anabolism
effect of testosterone on health- Pluymers RJ, Wouters EF (1995) in older women. J Clin Endocrinol
related quality of life in elderly Physiologic effects of nutritional Metab 91:38443849

204
Chapter 9: Androgen effects on the skeletal muscle

Shifren JL, Braunstein GD, Simon JA, Sinha-Hikim I, Roth SM, Lee MI, blind, placebo-controlled study.
Casson PR, Buster JE, Redmond Bhasin S (2003) Testosterone- J Clin Endocrinol Metab
GP, Burki RE, Ginsburg ES, Rosen induced muscle hypertrophy is 95:639650
RC, Leiblum SR, Caramelli KE, associated with an increase in Steidle C, Schwartz S, Jacoby K, Sebree
Mazer NA (2000) Transdermal satellite cell number in healthy, T, Smith T, Bachand R (2003)
testosterone treatment in women young men. Am J Physiol Endocrinol AA2500 testosterone gel normalizes
with impaired sexual function after Metab 285:E197E205 androgen levels in aging males with
oophorectomy. N Engl J Med improvements in body composition
Sinha-Hikim I, Taylor WE, Gonzalez-
343:682688 and sexual function. J Clin
Cadavid NF, Zheng W, Bhasin S
Sih R, Morley JE, Kaiser FE, Perry HM (2004) Androgen receptor in Endocrinol Metab 88:26732681
3rd, Patrick P, Ross C (1997) human skeletal muscle and cultured Storer TW, Magliano L, Woodhouse L,
Testosterone replacement in muscle satellite cells: up-regulation Lee ML, Dzekov C, Dzekov J,
older hypogonadal men: a 12- by androgen treatment. J Clin Casaburi R, Bhasin S (2003)
month randomized controlled Endocrinol Metab 89:52455255 Testosterone dose-dependently
trial. J Clin Endocrinol Metab increases maximal voluntary
82:16611667 Sinha-Hikim I, Cornford M, Gaytan H,
Lee ML, Bhasin S (2006) Effects of strength and leg power, but does
Singh R, Artaza JN, Taylor WE, testosterone supplementation on not affect fatigability or specific
Gonzalez-Cadavid NF, Bhasin S skeletal muscle fiber hypertrophy tension. J Clin Endocrinol Metab 88:
(2003) Androgens stimulate and satellite cells in community- 14781485
myogenic differentiation and dwelling older men. J Clin Storer TW, Woodhouse L, Magliano L,
inhibit adipogenesis in C3H Endocrinol Metab 91:30243033 Singh AB, Dzekov C, Dzekov J,
10T1/2 pluripotent cells through an Bhasin S (2008) Changes in muscle
androgen receptor-mediated Snyder PJ, Peachey H, Hannoush P,
Berlin JA, Loh L, Lenrow DA, mass, muscle strength, and power
pathway. Endocrinology but not physical function are
144:50815088 Holmes JH, Dlewati A, Santanna J,
Rosen CJ, Strom BL (1999a) Effect related to testosterone dose in
Singh R, Artaza JN, Taylor WE, Braga of testosterone treatment on body healthy older men. J Am Geriatr Soc
M, Yuan X, Gonzalez-Cadavid NF, composition and muscle strength in 56:19911999
Bhasin S (2006) Testosterone men over 65 years of age. J Clin Strawford A, Barbieri T, Van Loan M,
inhibits adipogenic differentiation Endocrinol Metab 84:26472653 Parks E, Catlin D, Barton N, Neese
in 3T3-L1 cells: nuclear R, Christiansen M, King J,
translocation of androgen Snyder PJ, Peachey H, Hannoush P,
Hellerstein MK (1999a) Resistance
receptor complex with beta-catenin Berlin JA, Loh L, Holmes JH,
exercise and supraphysiologic
and T-cell factor 4 may bypass Dlewati A, Staley J, Santanna J,
androgen therapy in eugonadal men
canonical Wnt signaling to Kapoor SC, Attie MF, Haddad JG Jr,
with HIV-related weight loss: a
down-regulate adipogenic Strom BL (1999b) Effect of
randomized controlled trial. JAMA
transcription factors. Endocrinology testosterone treatment on bone
281:12821290
147:141154 mineral density in men over 65
years of age. J Clin Endocrinol Strawford A, Barbieri T, Neese R, Van
Singh R, Bhasin S, Braga M, Artaza JN, Metab 84:19661972 Loan M, Christiansen M, Hoh R,
Pervin S, Taylor WE, Krishnan V, Sathyan G, Skowronski R, King J,
Sinha SK, Rajavashisth TB, Jasuja R Snyder PJ, Peachey H, Berlin JA, Hellerstein M (1999b) Effects of
(2009) Regulation of myogenic Hannoush P, Haddad G, Dlewati A, nandrolone decanoate therapy in
differentiation by androgens: cross- Santanna J, Loh L, Lenrow DA, borderline hypogonadal men with
talk between androgen receptor/b- Holmes JH, Kapoor SC, Atkinson HIV-associated weight loss. J Acquir
catenin and follistatin/TGF-b LE, Strom BL (2000) Effects of Immune Defic Syndr Hum Retrovirol
signaling pathways. Endocrinology testosterone replacement in 20:137146
150:12591268 hypogonadal men. J Clin Endocrinol
Metab 85:26702677 Tenover JL (1999) Testosterone
Sinha-Hikim I, Artaza J, Woodhouse L, replacement therapy in older adult
Gonzalez-Cadavid N, Singh AB, Lee Srinivas-Shankar U, Roberts SA, men. Int J Androl 22:300306
MI, Storer TW, Casaburi R, Shen R, Connolly MJ, OConnell MD,
Bhasin S (2002) Testosterone- Adams JE, Oldham JA, Wu FC Tenover JL (2000) Experience with
induced increase in muscle size in (2010) Effects of testosterone on testosterone replacement in the
healthy young men is associated muscle strength, physical function, elderly. Mayo Clin Proc 75 (Suppl):
with muscle fiber hypertrophy. Am body composition, and quality of S77S81; discussion:S82
J Physiol Endocrinol Metab 283: life in intermediate-frail and frail Tenover JS (1992) Effects of
E154E164 elderly men: a randomized, double- testosterone supplementation in the

205
Chapter 9: Androgen effects on the skeletal muscle

aging male. J Clin Endocrinol Metab composition in elderly men. J Clin N (2000) Transdermal testosterone
75:10921098 Endocrinol Metab 85:32763282 gel improves sexual function, mood,
Travison TG, Basaria S, Storer TW, Wagner GJ, Rabkin JG, Rabkin R muscle strength, and body
Jette AM, Miciek R, Farwell WR, (1998) Testosterone as a treatment composition parameters in
Choong K, Lakshman K, Mazer NA, for fatigue in HIV+ men. Gen Hosp hypogonadal men. Testosterone Gel
Coviello AD, Knapp PE, Ulloor J, Psychiatry 20:209213 Study Group. J Clin Endocrinol
Zhang A, Brooks B, Nguyen AH, Metab 85:28392853
Wang C, Eyre DR, Clark R, Kleinberg
Eder R, Lebrasseur N, Elmi A, D, Newman C, Iranmanesh A, Woodhouse LJ, Reisz-Porszasz S,
Appleman E, Hede-Brierley L, Veldhuis J, Dudley RE, Berman N, Javanbakht M, Storer TW, Lee M,
Bhasin G, Bhatia A, Lazzari A, Davidson T, Barstow TJ, Sinow R, Zerounian H, Bhasin S (2003)
Davis S, Ni P, Collins L, Bhasin S Alexander G, Swerdloff RS (1996a) Development of models to
(2011) Clinical meaningfulness of Sublingual testosterone replacement predict anabolic response to
the changes in muscle performance improves muscle mass and strength, testosterone administration in
and physical function associated decreases bone resorption, and healthy young men. Am J Physiol
with testosterone administration in increases bone formation markers Endocrinol Metab 284:
older men with mobility limitation. in hypogonadal men a clinical E1009E1017
J Gerontol A Biol Sci Med Sci research center study. J Clin Woodhouse LJ, Gupta N, Bhasin M,
66:10901099 Endocrinol Metab 81: Singh AB, Ross R, Phillips J, Bhasin
Urban RJ, Bodenburg YH, Gilkison C, 36543662 S (2004) Dose-dependent effects of
Foxworth J, Coggan AR, Wolfe RR, Wang C, Alexander G, Berman N, testosterone on regional adipose
Ferrando A (1995) Testosterone Salehian B, Davidson T, McDonald tissue distribution in healthy young
administration to elderly men V, Steiner B, Hull L, Callegari C, men. J Clin Endocrinol Metab
increases skeletal muscle strength Swerdloff RS (1996b) Testosterone 89:718726
and protein synthesis. Am J Physiol replacement therapy improves Yesalis CE, Bahrke MS (1995)
269:E820E826 mood in hypogonadal men a Anabolic-androgenic steroids.
van den Beld AW, de Jong FH, clinical research center study. J Clin Current issues. Sports Med
Grobbee DE, Pols HA, Lamberts Endocrinol Metab 81: 19:326340
SW (2000) Measures of bioavailable 35783583 Yesalis CE, Kennedy NJ, Kopstein AN,
serum testosterone and estradiol Wang C, Swerdloff RS, Iranmanesh A, Bahrke MS (1993) Anabolic-
and their relationships with muscle Dobs A, Snyder PJ, Cunningham G, androgenic steroid use in the United
strength, bone density, and body Matsumoto AM, Weber T, Berman States. JAMA 270:12171221

206
Chapter
Testosterone and cardiovascular disease

10 Kevin S. Channer and T. Hugh Jones

10.1 Epidemiology 207 10.8 Treatment studies in men with


10.2 Is estrogen cardio-protective? 208 atherosclerosis 217
10.3 Is testosterone cardio-protective? 208 10.8.1 Angina pectoris 217
10.4 Low testosterone and cardiovascular 10.9 Testosterone and heart failure 218
events 209 10.9.1 Pathophysiology 220
10.5 Low testosterone and accelerated 10.9.2 Androgen status in heart failure 220
atherosclerosis 209 10.9.3 Theoretical basis for testosterone as a
10.5.1 Coronary atheroma 209 treatment for heart failure 220
10.5.2 Carotid artery atheroma 210 10.9.4 Cardiac effects of testosterone
10.5.3 Peripheral arterial disease 210 treatment 221
10.6 Testosterone in animal models of 10.9.5 Non-cardiac effects of testosterone
atherosclerosis 211 treatment 221
10.7 What is the evidence for accelerated 10.9.6 Insulin resistance 221
atherosclerosis in hypogonadal states? 212 10.9.7 Inflammation 222
10.7.1 Effects on serum cytokine levels 213 10.9.8 Hemodynamics in heart failure 222
10.7.2 Effects on adhesion molecule 10.9.9 Erythropoietic effects of
expression 213 testosterone 222
10.7.3 Effects on vascular reactivity 214 10.9.10 Clinical trials of testosterone therapy
10.7.4 Effects on hemostatic factors 215 in heart failure 222
10.7.5 Effects on erythropoiesis 216 10.10 Testosterone levels and mortality 224
10.7.6 Effects on total and LDL 10.11 Key messages 226
cholesterol 216 10.12 References 226

10.1 Epidemiology populations; more than twice as many men are


Coronary heart disease remains the most important affected compared to women. This relationship per-
cause of death in the western world, and is increas- sists at all ages, so that at any age coronary death rates
ingly prevalent in the developing world because of are higher in men than women. This male predomin-
changes in risk factor profile. However, there remain ance is not explained by differences in the standard
large differences in the prevalence of ischemic heart risk factor profiles between men and women. Data
disease between different geographical areas of the from the British Heart Foundation show that there is
world. For example, Japan has a low prevalence and little difference between the frequencies of smoking,
Scotland a high prevalence (www.heartstats.org; hypertension, diabetes and hypercholesterolemia
Fig. 10.1). Despite these differences there is one epi- between men and women in the UK population.
demiological fact that is consistent between The observed difference in gender prevalence of

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

207
Chapter 10: Testosterone and cardiovascular disease

Belarus
Ukraine
Kazakstan
Azerbaijan
Russian Federation
Krygyzstan
Lithuania
Hungary
Romania
Czech Republic
lreland
Finland
UK
New Zealand
Austria
USA
Norway
Sweden
Denmark
Australia
Greece
Netherlands
Luxembourg Men
Italy
Spain Women
France
Japan
Fig. 10.1 Figure showing the consistent sex difference in death rates for coronary artery disease across both high- and low-prevalence
communities. Data are for men and women aged 3574 for 1998, from the BHF coronary statistics database released in 2002. See plate section
for color version.

coronary heart disease led to two theories that offset by a reduction in events in the latter years (years
females are in some way protected by virtue of their four and five) of the trial, leading the investigators to
sex hormone profile or that testosterone exerts a consider whether, in the long term, female combination
detrimental influence on the cardiovascular system. HRT is beneficial. An open-label, three-year follow-up
study (Grady et al. 2002) of the participants in the HERS
10.2 Is estrogen cardio-protective? study, which included 93% of the original cohort, also
showed no difference in cardiovascular events. These
Epidemiologically, coronary events are very rare in pre-
data show that HRT does not reduce the risk of further
menopausal women and only increase after the meno-
cardiovascular events in women.
pause, being more common in those with an early
A second large, randomized controlled trial (Ros-
menopause. Experimental data in animals do suggest
souw et al. 2002), of combination estrogen and proges-
that estrogens reduce the development of atheroscler-
togen versus placebo in over 16 000 postmenopausal
osis (see later). Similarly, coronary plaque burden has
women aged between 50 and 79 years, the Womens
been shown to be lower in women who use hormone
Health Initiative (WHI) study, was stopped early after
replacement therapy and in those who are taking the
5.2 years because of an excess of breast cancers. The data
oral contraceptive pill. This led to the widespread view
showed an excess cardiovascular risk although there was
that female hormones were protective and led to studies
no overall difference in all-cause mortality.
of hormone replacement therapy in postmenopausal
These clinical trial data have categorically demon-
women with the expectation that this would reduce
strated that female HRT is not cardio-protective. But
the incidence of cardiovascular disease.
what about testosterone in men?
However, two large randomized placebo-controlled
clinical trials did not show this. In the HERS study 10.3 Is testosterone cardio-
(Hulley et al. 1998), postmenopausal women,
of average age 66.7 years with a previous history of protective?
coronary heart disease, were randomly assigned to com- High doses of exogenous anabolic steroids are
bination estrogen and progestogen treatment or pla- undoubtedly associated with cardiac disease, sudden
cebo. Over the four years of the study there was no cardiac death and liver disease, but there is no evi-
difference in the incidence of cardiovascular events dence that high endogenous testosterone levels,
between the groups. However, in the first year of treat- within the normal range, are harmful. More import-
ment there was an excess of thrombotic events attrib- antly, coronary atherosclerosis increases with age
uted to the thrombogenic effects of estrogens. This was coincident with a fall in serum testosterone levels

208
Chapter 10: Testosterone and cardiovascular disease

(Simon et al. 1992). This in itself suggests that testos- It would be expected, therefore, that population-
terone does not induce atheroma formation. based follow-up studies would also show that a low
prevailing testosterone level is a marker of an
increased risk of cardiovascular events. The literature
10.4 Low testosterone and on this has been previously reviewed up to 2001 by
Wu and von Eckardstein (2003). At that time there
cardiovascular events were 39 studies which examined the relationship
There is one clinical scenario where the effect of a reduc- between testosterone blood level and coronary artery
tion in circulating testosterone on cardiac disease can be disease (CAD). Of 32 cross-sectional studies, half
seen. In men with prostate cancer, androgen deprivation found that subjects with CAD had lower levels of
therapy (ADT), including castration, has been used for testosterone than controls, and half found no differ-
years for palliation. In 1998, Satariano et al. (1998) first ence. Of the seven prospective, nested, case control
reported the observation that cardiovascular deaths were studies, none showed that low testosterone was asso-
second only to death from the cancer in these men. The ciated with an increased risk of developing overt
size of the excess cardiovascular detriment in this setting symptomatic CAD.
is significant. In one observational study of 73 196 men This inconsistency can be explained by a number
aged 66 years or older with prostate cancer, followed for of factors. There is a biological and diurnal variation
10 years, those treated with androgen deprivation ther- in testosterone blood level which none of these studies
apy experienced a 44% increase in the risk of diabetes and took into account, as most were not conducted spe-
a 16% increase in the risk of coronary heart disease events cifically to address this question. All the prospective
(death or myocardial infarction) (Keating et al. 2006). studies looked at a number of risk factors, and testos-
Orchidectomy was associated with a 34% increase in terone blood level was often an afterthought. The
diabetes but no increased risk of coronary heart disease. influence of the timing of sampling, type of assay,
In a study of over 22 000 men with prostate cancer, those storage, and effect of co-morbidities on testosterone
who received ADT had a 20% increased risk of cardio- blood level effectively dilutes the power of these stud-
vascular morbidity in the first year of treatment (Saigal ies. More importantly, it is increasingly recognized
et al. 2007) compared with those who did not. Another that the sensitivity of the AR is very important in
study showed that men over the age of 65 years treated translating the biological effects of the prevailing tes-
with ADT had a two-fold increased risk of a fatal cardio- tosterone blood level. Without knowledge of the
vascular event over a 10-year follow-up period (Tsai receptor sensitivity in individuals it is very difficult
et al. 2007). Several studies have now shown that ADT to understand the biological consequence of a particu-
causes increased cardiovascular co-morbidity, probably lar level of testosterone in the circulating blood.
through an increase in insulin resistance (reviewed in An alternative method of assessing the influence
Shahani et al. 2008 and Taylor et al. 2009), which is an of testosterone on atherosclerosis is to use prospective
associated consequence of hypotestosteronemia. There cohort studies of actual markers of atherosclerosis,
are studies which have not shown an effect of cardiovas- with individuals acting as their own controls. These
cular mortality, like the small EORTC (European Organ- studies are all consistent in demonstrating that low
isation for Research and Treatment of Cancer) 226863 testosterone in an individual increases atherogenesis.
trial (Bolla et al. 2010), but the totality of the evidence has
led to a science advisory statement from the American
Heart Association, American Cancer Society and the 10.5 Low testosterone and
American Urological Association to recommend that accelerated atherosclerosis
all patients receiving ADT have cardiovascular risk factor There are a number of observational models of ather-
assessment on a regular basis, and that those with coexist- oma development in humans.
ent cardiovascular disease have their secondary preven-
tion treatment optimized (Levine et al. 2010).
These studies show that manipulation of the indi- 10.5.1 Coronary atheroma
vidual prevailing hormonal profile to produce a hypo- There have been a number of relatively small studies
gonadal state results in an excess of cardiovascular of the relationship between CAD and testosterone
events and death. blood level, dating from 1985. Some have been

209
Chapter 10: Testosterone and cardiovascular disease

positive (Chute et al. 1987; English et al. 2000a; Dobr- However, in a further study from the same group,
zycki et al. 2003; Rosano et al. 2007; Malkin et al. there was no correlation between the change in plaque
2010), and some negative (Hauner et al. 1991; area, measured seven years apart in over 1000 men,
Kabakci et al. 1999; Davoodi et al. 2007). Phillips and testosterone levels (Vikan et al. 2009). By com-
et al. (1994) published the first study, of men with parison, two other serial studies have shown that men
CAD identified at coronary angiography, to demon- with lower levels of testosterone develop a greater
strate a relationship between the prevailing free increase in the intima-media thickness over time,
testosterone blood level and the degree of athero- indicating accelerated atherogenesis (Muller et al.
sclerosis. The findings showed a negative correlation 2004). Men with clinically significant carotid artery
even after adjusting for body mass index and age. stenosis also appear to have lower than normal levels
Moreover, these authors also demonstrated later of testosterone. In a case control study, 124 men
(Phillips et al. 2004) that standard accepted cardio- having carotid endarterectomy had significantly lower
vascular risk factors (smoking, hypertension, dia- total testosterone and SHBG levels than controls
betes, hypercholesterolemia and obesity) did not (Debing et al. 2008). A multivariate analysis including
closely correlate with the degree of atherosclerotic relevant clinical and physiological factors showed a
coronary artery stenosis at angiography, whereas age, significant inverse correlation between total testoster-
free testosterone level and HDL-cholesterol level did. one and the degree of internal carotid artery stenosis.
A large study of 930 men with angiographically In one small treatment study, carotid intima-
proven coronary artery disease found that 16.3% had media thickness appeared to be reduced following
total testosterone levels < 8.1 nmol/l and 20.9% had physiological replacement therapy in men with low
bioavailable testosterone below the normal range baseline testosterone levels (Mathur et al. 2009).
(<2.6 nmol/l) (Malkin et al. 2010). These interventional data need to be repeated in a
There are many potential confounding factors larger cohort to satisfactorily answer the question of
which influence testosterone level, including acute whether TRT does ameliorate the age-related progres-
inflammation and illness, technical assay differences sion of atherosclerosis in men.
and the relative importance of bioavailable testoster-
one compared with total testosterone. In a case control
study of men having coronary angiography, total and 10.5.3 Peripheral arterial disease
bioavailable testosterone was significantly lower in Similarly, atherosclerosis identified as calcified aortic
men with CAD compared to those with normal arter- plaques on lateral abdominal X-rays was shown to be
ies (English et al. 2000a). In this careful study men significantly more advanced in individuals who had a
were excluded from the study if they suffered an acute low testosterone level over a follow-up period of seven
illness or acute infarction within the previous three years (Hak et al. 2002). Thoracic aortic intima-media
months, and blood samples were taken in the early thickness has been studied by trans-esophageal echo-
morning period (before 08.3009.30). Moreover, the cardiography in a small study of 42 men who had no
strongest association was seen with bioavailable tes- clinical evidence of vascular disease (Demirbag et al.
tosterone rather than either total or free testosterone. 2005). These authors demonstrated an inverse rela-
tionship between total testosterone level and aortic
intima-media thickness.
10.5.2 Carotid artery atheroma Atherosclerosis in another vascular bed has been
Carotid atherosclerosis as identified by an increase in studied. Retinal atherosclerosis has been associated
the intima-media thickness in the carotid artery, with a low DHEA blood level but not with a low total
using Doppler ultrasound scanning, is more common testosterone level (Tedeschi-Reiner et al. 2009). In a
in men with a lower level of testosterone (Makinen Swedish cross-sectional study involving nearly 3000
et al. 2005). In a large cross-sectional population men, a multivariate analysis showed that low free
study in Sweden of over 1400 men (Svartberg et al. testosterone and high free estradiol were independ-
2006), there was a negative correlation between ently associated with a reduction in ankle-brachial
carotid intima-media thickness and total testosterone index, which is a marker of peripheral vascular dis-
level, even after adjustment for age, smoking, systolic ease (Tivesten et al. 2007). Arterial stiffness of large
blood pressure and use of lipid-lowering medication. arteries is a surrogate marker for atherosclerosis and

210
Chapter 10: Testosterone and cardiovascular disease

can be measured non-invasively by the carotid-femoral no treatment. Testosterone administration signifi-


pulse wave velocity. In a cross-sectional study of 376 cantly inhibited atheroma formation in male rabbits,
subjects, a low pulse wave velocity was inversely related whilst an increase in plaque size was noted in females;
to total and bioavailable testosterone levels, although no protective effect was seen in male rabbits given
the statistical strength of the association was lost when estradiol, although it significantly inhibited atheroma
age was included (Pour et al. 2007). In a recent Japan- in females. Interestingly, simultaneous administration
ese study, however, although low testosterone blood of both testosterone and estradiol reduced atheroma
level predicted cardiovascular events it appeared to be formation in both sexes. Alexandersen et al. (1999)
independent of changes in flow-mediated brachial examined the effects of testosterone and DHEA on
reactivity (Akishita et al. 2010). atheroma formation in sexually mature male rabbits.
Men with end-stage kidney disease on mainten- Rabbits were randomized to bilateral castration or
ance hemodialysis have a high incidence of hypogo- sham operation, with the castrated animals subse-
nadal levels of blood testosterone. In one study of 100 quently assigned to four study groups: (1) 500 mg oral
non-diabetic men of whom half had low blood levels DHEA daily, (2) 80 mg oral testosterone undecanoate
of testosterone, vascular reactivity was reduced and daily; (3) 25 mg im injection of testosterone
carotid intima-media thickness increased compared enanthate, twice weekly; or (4) placebo. The sham-
to men with normal testosterone levels (Karakitsos operated group received no treatment. Animals were
et al. 2006). fed a pro-atherogenic diet for 35 weeks to induce
The mechanism underlying the observed excess aortic atherosclerosis. Alexandersen et al. reported a
cardiovascular events may be explained by accelerated doubling of aortic plaque formation in castrated com-
atherosclerosis as a consequence of changes in the pared to sham-operated animals. Treatment with oral
cardiovascular risk factor profile that occur as testos- testosterone or DHEA prevented this increase in
terone blood level falls. atheroma formation; whilst treatment with im testos-
terone was associated with a significant reduction in
10.6 Testosterone in animal models plaque formation. Others also report that DHEA
inhibits plaque formation in this model (Gordon
of atherosclerosis et al. 1988; Arad et al. 1989; Eich et al. 1993); whilst
There are a number of animal models showing that testosterone is also reported to inhibit neo-intimal
castration or hypogonadism is associated with plaque development in endothelial denuded rabbit
increased atherosclerosis. Moreover, supplemental aortic rings (Hanke et al. 2001).
testosterone therapy to physiological levels amelior- Physiological testosterone supplementation has
ates or prevents the aortic accumulation of choles- also been shown to reduce atherosclerosis in orchid-
terol, with a reduction in plaque size in models of ectomized LDL receptor knock-out mice fed a choles-
atherosclerosis. Larsen et al. (1993) published the first terol-enriched diet for a period of eight weeks
study examining the effects of testosterone replace- (Nathan et al. 2001). Increased aortic lesion forma-
ment in orchidectomized male rabbits fed a pro- tion was reported in orchidectomized male mice;
atherogenic diet. Animals received supplemental whilst a reduction in plaque development was dem-
testosterone enanthate (100 ml of 250 mg/ml concen- onstrated in orchidectomized animals receiving
trated solution), for a period of 17 weeks via twice- physiological testosterone supplementation. However,
weekly im injection. Following cholesterol feeding, this effect was not observed in animals treated simul-
aortic cholesterol accumulation was significantly taneously with testosterone and the aromatase inhibi-
retarded in the testosterone-treated, compared to the tor anastrozole. In contrast to Bruck et al. (1997), this
placebo-treated, group (Larsen et al. 1993). Compar- study demonstrated that conversion to estradiol was
able to these findings, Bruck et al. (1997) demon- responsible for the testosterone-mediated attenuation
strated gender-specific anti-atherogenic effects of of lesion formation. However, more recently Qui et al.
testosterone and estradiol in orchidectomized male (2010) showed that the non-aromatized natural
and female rabbits fed a pro-atherogenic diet for a androgen 5a-dihydrotestosterone (DHT), when used
12-week period. Rabbits received either testosterone as androgen replacement in castrated rabbits, reduced
enanthate (25 mg/kgwk) or estradiol (1 mg/kgwk), the fatty streak development in the aorta by reducing
alone or in combination via weekly im injection, or foam cell formation from macrophages.

211
Chapter 10: Testosterone and cardiovascular disease

These studies show that the mechanism underpin- In a castrated rabbit model (Li et al. 2008), aortic
ning the effect on atherosclerosis remains controver- plaque growth was reduced by TRT, but this effect
sial. Does testosterone exert its anti-atherosclerotic was lost when flutamide (a nuclear AR blocker) was
effect through conversion to estrogen, or is it an given concurrently. Castration increased inflamma-
independent effect? Another controversial area con- tory cytokine levels; an effect blocked by TRT but
cerns the relative importance of the nuclear AR in the only in the absence of flutamide. These data also
mechanism of this effect. suggest that the nuclear AR is important in modulat-
In one animal model of hypogonadism in mice, in ing the effect of testosterone on atheroma formation.
which the blood testosterone level is only 10% of litter-
mate controls, and which also has an absent nuclear
AR (the testicular feminized mouse), physiological 10.7 What is the evidence for
testosterone supplementation reduces fatty streak accelerated atherosclerosis in
formation (Nettleship et al. 2007a). This study utilized
Tfm (testicular feminized) mice (n = 31) and XY lit- hypogonadal states?
termates (n = 8) which were separated into five experi- In order to better understand how changes in testos-
mental groups. Each group received saline (Tfm, n = 8; terone blood level may influence vascular function
XY littermates, n = 8), physiological testosterone alone and structure, it is necessary to understand the patho-
(Tfm, n = 8), physiological testosterone in conjunction physiology of atherosclerosis.
with the ERa antagonist fulvestrant (Tfm, n = 8), or Atherosclerosis is a disease of the intima of the
physiological testosterone in conjunction with the medium and large arteries including the aorta, carotid
aromatase inhibitor anastrozole (Tfm, n = 7). All and cerebral arteries. Coronary artery disease is char-
groups were fed a cholesterol-enriched diet for 28 acterized pathologically by the atherosclerotic plaque,
weeks. Serial sections from the aortic root were exam- which is described as a focal inflammatory fibro-pro-
ined for fatty streak formation, and whole blood liferative response to multiple forms of endothelial
was collected for measurement of total cholesterol, injury, in which a number of distinct but overlapping
high-density lipoprotein cholesterol (HDL-C), non- pathways of pathogenesis are involved.
HDL-C, testosterone and 17b-estradiol. This study It is thought that the principal event initiating
demonstrated that physiological testosterone replace- atheromatous lesion formation is endothelial cell
ment significantly reduced fatty streak formation in injury. This encourages monocyte attachment, via
Tfm mice compared with placebo-treated controls; an the expression of adhesion molecules and chemokines
effect that was independent of the androgen receptor. on the endothelial cell surface. When activated by
The effect persisted even in the presence of an ER injury, endothelial cells and the attached monocytes
blocker and an aromatase inhibitor. Serum HDL-C and macrophages generate free radicals, which oxidize
was also significantly increased following physiological LDL, resulting in lipid peroxidation and destruction of
testosterone replacement; however co-treatment with the receptor needed for normal receptor-mediated
either fulvestrant (an ER blocker) or anastrozole (an clearance of LDL. Consequently, oxidized LDL (ox-
aromatase inhibitor) demonstrated that the observed LDL) accumulates in the sub-endothelial space, where
increase in HDL-C was consistent with conversion to it is taken up by macrophages via scavenger receptors
17b-estradiol (Nettleship et al. 2007a). to form foam cells (Ross 1999). Oxidized LDL is pro-
In an AR knock-out, apolipoprotein E deficient posed to exacerbate the local inflammatory response,
mouse model which is also testosterone deficient, and has other effects, such as inhibiting the produc-
Bourghardt et al. (2010) also showed that fatty streak tion of nitric oxide, an important chemical mediator
development was increased when they were fed a high with multiple anti-atherogenic properties, including
lipid diet, and this effect was reduced by the addition vasorelaxation. After taking up LDL, these macro-
of testosterone. They also showed that fatty streak was phages (now foam cells) migrate sub-endothelially.
increased in orchidectomized mice and reduced with Sub-endothelial collections of foam cells and to a
testosterone replacement. The authors suggested that lesser extent T-lymphocytes form the fatty streaks,
these results implied that low testosterone states are which presage atherosclerosis.
associated with accelerated atherosclerosis by both an Although the fatty streaks themselves are not
AR dependent and independent mechanisms. clinically significant, they are now accepted to be the

212
Chapter 10: Testosterone and cardiovascular disease

precursors of more advanced lesions, which may go Testosterone appears capable of modulating both
on to become the sites of thrombosis (Lusis 2000). pro- and anti-inflammatory cytokine production by
Advanced atheroma is characterized by the leukocytes in vitro (Li et al. 1993; Kanda et al. 1996;
following histological criteria: accumulations of lipid, 1997; Bebo et al. 1999; DAgostino et al. 1999; Liva and
SMC, T-lymphocytes, macrophages, necrotic cell Voskuhl 2001; Corrales et al. 2006). Castrated male
debris and matrix components (including minerals), mice are reported to have increased TNF-a production,
associated with structural disorganization and following administration of bacterial endotoxin; an
thickening of the intima, with deformity of the arter- effect which was suppressed by subsequent testosterone
ial wall. The advanced lesion is contained within a replacement (Spinedi et al. 1992). Similarly, young hypo-
fibrous cap, formed by SMC proliferation and vascu- gonadal men with delayed puberty caused by IHH are
lar remodeling, the result of the unabated response to reported to exhibit elevated serum levels of inflammatory
endothelial injury. The cap consists of SMC sur- cytokines compared to healthy controls, which can be
rounded by collagen, elastic fibers and proteoglycans corrected by TRT (Yesilova et al. 2000). Elderly, hypo-
(Tedgui and Mallat 2001). gonadal men have also been found to have raised serum
Rupture of the fibrous cap results in the spillage of levels of TNF-a and interleukin 6 (Khosla et al. 2002).
the contents of the advanced lesion into the vessel Testosterone replacement therapy reduces cyto-
lumen. This results in the formation of a thrombus, kine activation in hypogonadal men. Malkin et al.
which either heals, causing further lesion progression (2004a) completed a randomized, single-blind, pla-
and further compromising flow at the local site, or cebo-controlled crossover study of physiological tes-
occludes the coronary artery completely resulting in tosterone therapy (one month of Sustanon100 via
myocardial infarction and sudden death. Plaque fortnightly im injection) in 10 men with CAD and
rupture and thrombosis are the primary complica- hypotestosteronemia. This level of TRT was sufficient
tions of the advanced lesion, which cause unstable to significantly reduce serum levels of TNF-a. The
coronary syndromes (Ross 1999). investigators repeated their work in a larger study
Over recent years increasing evidence has emerged using the same testosterone dosing regimen, but using
that a number of the cellular mechanisms intimate to a randomized, placebo-controlled crossover protocol
the atherosclerotic process are modulated beneficially in 27 hypogonadal men with concomitant cardiovas-
by testosterone. cular disease (Malkin et al. 2004b). Physiological TRT
was again shown to significantly lower serum levels of
TNF-a, and also induced a significant decrease in
10.7.1 Effects on serum cytokine levels serum levels of IL-1b
As well as the reduction in
Cytokines are key players in the atherosclerotic process. pro-inflammatory cytokines following physiological
Pro-inflammatory cytokines such as interleukin 1-beta testosterone administration, a significant elevation
(IL-1b) and tumor necrosis factor alpha (TNF-a) are was observed in serum levels of the anti-inflammatory
known to increase adhesion molecule expression, pro- cytokine interleukin 10 (IL-10).
mote SMC proliferation and induce matrix metallopro-
teinase activity, which are critical mechanisms in the
initiation of plaque development and rupture. Observa-
10.7.2 Effects on adhesion molecule
tional evidence suggests that inflammatory cytokines expression
and serum testosterone levels are interconnected. In Testosterone is reported to have a modulatory effect
one study of men with CAD, it was shown that serum upon endothelial cell adhesion molecule expression.
levels of IL-1b increased significantly in a step-wise Dihydrotestosterone has been shown to increase
manner in line with the atherosclerotic burden (Nettle- monocyte adhesion to IL-1b-stimulated human
ship et al. 2007b). Moreover, when patients were classi- umbilical vein endothelial cells (HUVECs) and human
fied by their serum testosterone level, being either umbilical artery endothelial cells (HUVACs) obtained
eugonadal, borderline hypogonadal or hypogonadal, from male donors, and also to increase IL-1b-induced
a step-wise increase in IL-1b with lower testosterone vascular cell adhesion molecule 1 (VCAM-1) expres-
status was observed. These data suggest that the under- sion in these cells (McCrohon et al. 1999), via acti-
lying testosterone status may modulate IL-1b produc- vation of nuclear factor kappa B (NF-kB) transcription
tion in men with CAD. factor. Similarly, Zhang et al. (2002) reported an

213
Chapter 10: Testosterone and cardiovascular disease

increase in TNF-a-induced expression of VCAM-1 (Yue et al. 1995; Chou et al. 1996; Webb et al. 1999a;
and the cell adhesion molecule (E-selectin), which Crews and Khalil 1999a; English et al. 2000b; 2001;
mediates monocyte adhesion to cytokine-activated Deenadayalu et al. 2001; Pugh et al. 2002a; Jones et al.
endothelial cells, by testosterone in HUVECs. In both 2004a); an action which diminishes with aging
studies the action of testosterone was abolished by AR (English et al. 2000b). Testosterone-induced vasodila-
antagonism. In contrast, testosterone reduced TNF-a- tation is rapid in onset and is unaffected by AR block-
induced VCAM-1 expression in HUVECs of female ade (Yue et al. 1995; English et al. 2000b; Tep-areenan
origin, an activity blocked by ER antagonism and et al. 2002; Jones et al. 2002; 2004b) or deficiency
aromatase inhibition (Mukherjee et al. 2002). These (Jones et al. 2003a), and is preserved in endothelial-
data suggest that testosterone may have a detrimental denuded vessels (Perusquia et al. 1996; Perusquia and
influence upon adhesion molecule expression in males Villalon 1999; Murphy and Khalil 1999; Crews and
via an interaction with the AR, but induces potentially Khalil 1999b; Honda et al. 1999) and in the presence
beneficial reductions in adhesion molecule expression of nitric oxide synthase inhibitors (Yue et al. 1995;
in females via aromatization to 17b-estradiol. Honda et al. 1999; Deenadayalu et al. 2001; Jones
However, vascular specificity is also apparent. et al. 2004a; 2004b), guanylate cyclase or cyclooxygen-
Importantly, testosterone is reported to reduce TNF- ase (Yue et al. 1995; Chou et al. 1996; Jones et al. 2004a;
a-induced VCAM-1 expression in human aortic 2004b; Crews and Khalil 1999a; 1999b). These obser-
endothelial cells (Hatakeyama et al. 2002). This cell vations clearly demonstrate that the vasodilatory
line is more relevant in terms of atherosclerosis, com- action of testosterone is mediated directly via an inter-
pared to umbilical cells. Evidently further study is action at the level of the vascular smooth muscle.
warranted, but these data suggest that testosterone Furthermore, the observation that testosterone-
could potentially exert a beneficial effect upon adhe- induced dilatation is not attenuated by covalent link-
sion molecule expression. age to albumin, which prevents its endocytosis into
Neo-intimal proliferation occurs after vessel wall the SMC (Ding and Stallone 2001; Jones et al. 2004a),
injury, for example during angioplasty, and is a cause implies that the dilatory signaling process is initiated
for early re-stenosis. One novel study in pigs, which at the smooth muscle cell membrane, which has been
have anatomically and functionally similar coronary demonstrated to contain testosterone binding sites
arteries to humans, has shown that testosterone also (Jones et al. 2003b; 2004a; 2004b). The proposed
influences vascular repair mechanisms. In this study, underlying mechanism of action of testosterone is
pigs were castrated and then either given TRT or thought to occur via activation of calcium-sensitive
placebo. Those pigs given TRT had reduced neo-inti- potassium channels (KCa) (Deenadayalu et al. 2001;
mal proliferation compared to those with a prevailing Tep-areenan et al. 2002). Alternatively a number of
low testosterone level (Tharp et al. 2009). By com- studies report an agonist-dependent variance in the
parison, when exogenous additional testosterone or vasodilatory efficacy of testosterone (Yue et al. 1995;
dihydrotestosterone is given to normal animals, arter- Crews and Khalil 1999a; 1999b; Perusquia and Villa-
ial calcification is increased (McRobb et al. 2009). lon 1999; Murphy and Khalil 1999; Ding and Stallone
2001; English et al. 2002; Jones et al. 2002), consistent
with calcium antagonistic action upon voltage-gated
10.7.3 Effects on vascular reactivity calcium channels (VGCCs) (Jones et al. 2003c),
Maintenance of a correct response to vasoconstrictive although an inhibitory action upon store-operated
and vasodilatory agents is essential in the control of calcium channels (SOCCs) also occurs in the systemic
vascular tone. This is especially important in athero- vasculature (Jones et al. 2003a). Endogenous testos-
sclerosis within the coronary circulation, where terone has been shown to increase the expression of
reduced vasodilatation and enhanced vasoconstriction L-type calcium channels by a direct action on the gene
causes further restriction of coronary blood flow promoter in porcine coronary smooth muscle cells
through the partially occluded atherosclerotic vessel, (Bowles et al. 2004). Electrophysiological studies have
and can also lead to vasospasm, thereby exacerbating demonstrated that testosterone in physiological con-
anginal symptoms. In both human and animals centrations inhibits both L-type and T-type VGCCs in
models testosterone has been shown to elicit marked vascular SMC (Scragg et al. 2004). These findings are
coronary vasodilatation, both in vivo and in vitro supported by microfluorimetric studies in the rat

214
Chapter 10: Testosterone and cardiovascular disease

smooth muscle cell line A7r5 which showed that DHT Hypotestosteronemia is associated with hyperten-
as well as testosterone inhibited the L-type calcium sion and arterial stiffening. Androgen deprivation
channel (Hall et al. 2006). The effects of physiological therapy for prostate carcinoma leads to increased
testosterone concentrations were absent in the pres- vascular stiffness assessed after three months (Smith
ence of nifedipine, an L-type calcium channel blocker, et al. 2001). There have been several trials of TRT in
but not the T-type calcium channel blocker pimozide. eugonadal, hypogonadal and obese men which have
This is likely to be mediated via direct binding to the observed impressive reductions in both systolic and
main a1C subunit of the VGCC, since a similar inhibi- diastolic blood pressure over periods as short as six
tory action is observed in HEK293 cells transfected months and for as long as 10 years (Mrin et al. 1993;
with this channel protein. This is supported by the Anderson et al. 1996; Zitzmann and Nieschlag 2007).
finding that a single point mutation at the nifedipine
binding site renders the effect of testosterone inactive
(Scragg et al. 2007). This evidence strongly demon- 10.7.4 Effects on hemostatic factors
strates that testosterone is a natural ligand for the In the majority of cases, myocardial infarction occurs
nifedipine binding site on L-type calcium channels. as a result of coronary thrombosis, triggered by ath-
Testosterone also beneficially modulates responses erosclerotic plaque rupture and disruption of the vas-
induced by other vasoactive stimuli. Both chronic cular endothelium. The thrombotic process is
exposure to physiological testosterone therapy (Kang complex, and dependent upon a variety of intrinsic
et al. 2002) and acute exposure to supraphysiological pro- and anti-thrombotic mediators which determine
doses of testosterone (Ong et al. 2000) are reported to the coagulation status. The anti-coagulation agents
increase flow-mediated brachial artery vasodilatation, tissue plasminogen activator (tPA) and tissue factor
occurring as a result of increased nitric oxide release pathway inhibitor (TFPI), and the pro-thrombotic
from the endothelium in response to changes in sheer factor plasminogen activator inhibitor 1 (PAI-1) are
stress in men with CAD. Long-term physiological tes- integral to this process. Plasminogen activator inhibi-
tosterone therapy also improves nitrate-mediated bra- tor 1 is a predictor of myocardial infarction and
chial artery vasodilatation in these patients (Kang et al. progression of atherosclerosis in patients with stable
2002). Brachial artery reactivity correlates closely with CAD (Thogerson et al. 1998; Bavenholm et al. 1998).
coronary arterial responsiveness (Anderson et al. Evidence suggests that low serum testosterone is asso-
1995a), which would support a beneficial long-term ciated with a hyper-coagulable state. Indeed, serum
effect for testosterone upon atherosclerotic coronary testosterone levels and tPA are reported to be posi-
vasomotion. By contrast, hypogonadal men without tively correlated, whilst a negative correlation exists
cardiovascular disease exhibit elevated flow- and between serum levels of testosterone and PAI-1 and
nitrate-mediated brachial artery vasodilation (Zitz- clotting factor VII (the levels of which are reduced by
mann et al. 2002), which is restored to control levels TFPI) (Glueck et al. 1993; Phillips et al. 1994; 1995;
following testosterone replacement. Similarly flow- Pugh et al. 2002b). Moreover, both testosterone
mediated, but not nitrate-mediated, brachial artery replacement in hypogonadal men, and androgen
vasodilatation has been shown to be increased in men treatment in healthy men, leads to reduced PAI-1
in whom testosterone levels have been therapeutically levels (Caron et al. 1989; Beer et al. 1996). Similarly,
or surgically lowered, as a treatment for prostate car- a number of cross-sectional studies have shown that
cinoma (Herman et al. 1997). Discrepancies may be hypotestosteronemia is associated with high levels of
due to the residual density of the ARs, which are the acute phase protein, fibrinogen (Yang et al. 1993;
upregulated in hypogonadal men in response to lower De Pergola et al. 1997), and that testosterone replace-
endogenous testosterone (Malkin et al. 2006a). In ment can reduce fibrinogen levels (Anderson et al.
circumstances where there is combined testosterone 1995b. This beneficial effect on coagulation status
and AR deficiency in the testicular feminized mouse, appears to be lost in men who have developed CAD,
there is reduced endothelial-dependent vasodilatation as physiological TRT in men with CAD has recently
(Jones et al. 2003a). These data implicate an interaction been reported to have no effect on serum fibrinogen
between testosterone acting at the cell membrane and levels, tPA or PAI-1 (Smith et al. 2005).
the nuclear AR in the long-term regulation of vascular In a long-term clinical study from Sweden (Svart-
tone (reviewed in Jones et al. 2004b). berg et al. 2009), of 1350 men followed up for 10 years,

215
Chapter 10: Testosterone and cardiovascular disease

androgen levels in men were not associated with risk of average, 1.42  1.55 g/dl, and hematocrit increased by
venous thromboembolism. This is an important 0.03  0.04 g/dl.
observation, since an increased risk of arterial and In long-term studies of injectable TRT, hemo-
venous thromboembolism is associated with both globin and hematocrit have been shown to increase,
female hormone replacement therapy (HRT) and use but not above the acceptable normal ranges (Minne-
of the oral contraceptive pill (WHO 1997). This mann et al. 2007).
is thought to account for the increased incidence in Although there is potential for adverse effects on
vascular events and death associated with HRT in the the circulation of a higher than normal hematocrit,
recent Womens Health Initiative study (Rossouw et al. there is also the potential for benefit. Chronic heart
2002; see above). This adverse effect on coagulation is failure is associated with persistent inflammatory acti-
not a property shared by testosterone. vation, and chronic anemia is part of the syndrome
especially in cachectic patients.

10.7.5 Effects on erythropoiesis


Testosterone treatment may increase erythropoiesis, 10.7.6 Effects on total and LDL cholesterol
and, as a consequence, increase hematocrit. There is a Elevated serum cholesterol, especially LDL choles-
dose-related effect on erythropoiesis which is more terol, is a powerful risk factor for the premature
striking in older rather than younger men (Coviello development of atherosclerosis. It is the sub-endothe-
et al. 2008). The effect is seen within a month of lial accumulation of LDL cholesterol that provokes a
replacement therapy and peaks by 12 months, but is local inflammatory response, that results in the gen-
not associated with a change in either erythropoietin eration of fatty streaks (smooth raised plaques
or soluble transferrin receptor (Coviello et al. 2008). located beneath the endothelium), which represent
The mechanism whereby testosterone increases ery- the initial phase of atherosclerosis.
thropoiesis remains unknown. This in itself may pose Low endogenous serum levels of testosterone have
a threat by theoretically increasing thrombotic risk. been reported, in a number of cross-sectional studies,
Guidelines recognize this potential issue and recom- to be correlated with higher serum levels of total
mend that when TRT is initiated that the hematocrit and LDL cholesterol (Barrett-Connor and Khaw 1988;
is monitored after 3 and 12 months and then annu- Barrett-Connor 1992; Haffner et al. 1993; Simon et al.
ally. It is suggested that treatment be interrupted if the 1997; Isidori et al. 2005; Mkinen et al. 2008). These
hematocrit rises above 55%, and/or venesection be data suggest that hypogonadal men may exhibit an
offered to maintain this level (Wang et al. 2008). adverse pro-atherogenic lipid profile. Moreover, TRT
Earlier studies suggested that this effect is seen causes significant reductions in total and LDL choles-
mainly when replacement therapy is given in high terol in hypogonadal men (Tenover 1992; Tripathy
dosage or when the mode of delivery exposes the et al. 1998; Howell et al. 2001; Ly et al. 2001); an effect
individual to transient high levels. which is maintained in eugonadal individuals (Thomp-
In the latest study of topical treatment with a 2% son et al. 1989; Bagatell et al. 1994; Uyanik et al. 1997).
gel applied daily to the axilla in androgen-deficient Serum total cholesterol levels have been shown to
men with symptoms of hypogonadism, the treatment decline following physiological testosterone replace-
was discontinued in four men (2.6%) after three ment in hypogonadal men with CAD, despite these
months of treatment because of a high hematocrit patients already being treated with statin drugs
above 0.54% (Wang et al. 2011). A previous study, (HMG-CoA reductase inhibitors) (Malkin et al. 2004a).
involving 227 men who used a 1% gel, had shown Although LDL cholesterol is associated with an
significant changes in hemoglobin or hematocrit, adverse cardiovascular profile, so HDL cholesterol is
with 11.3% exceeding the upper normal range for associated with a reduced risk profile. Testosterone
hematocrit at 50 mg/day and none at this dose has been shown to reduce HDL-C in some (Bagatell
needing to stop therapy (Wang et al. 2000). In et al. 1994; Rossouw et al. 2002), but not all
another large study of 220 men with type 2 diabetes (Gliczynski et al. 1996; Uyanik et al. 1997) studies.
or metabolic syndrome and hypogonadism (Jones However, reductions in HDL-C are generally smaller
et al. 2011), gel replacement therapy was associated and less pronounced than those of other lipid frac-
with a non-significant increase in hemoglobin of, on tions (Whitsel et al. 2001; Jones and Saad 2009).

216
Chapter 10: Testosterone and cardiovascular disease

In animal models, serum levels of total cholesterol showed clinical improvement with testosterone in
are raised in the testosterone-deficient testicular fem- 91%, with no change in 9%.
inized (Tfm) mouse. Furthermore, following testos- The first randomized controlled trial was con-
terone supplementation in the Tfm mouse, HDL-C ducted in 1977. Jaffe (1977) published the results of
was demonstrated to be significantly raised compared the effects of weekly injections of testosterone
to placebo controls (Nettleship et al. 2007a). cypionate or placebo on post-exercise ST segment
Triglycerides have also been implicated in athero- changes on the 12-lead electrocardiograph (ECG)
genesis, particularly in diabetic populations. In a large after the two-step test (an early form of exercise
study of middle-aged men with hypogonadal symp- testing). He showed statistically significant reductions
toms, low testosterone was associated with high tri- in the sum of ST segment change by 31.7% at the end
glycerides as well as low HDL-C (Mkinen et al. of four weeks and 51.2% by eight weeks of treatment.
2008). In one clinical study (Agledahl et al. 2008), The only other positive finding was an increase in
elderly men with low testosterone levels had higher hemoglobin concentration of 0.71.4 g/dl by the end
BMI, waist circumference and triglyceride levels after of the trial. However, Jaffe found no correlation
fatty meals. between the change in hemoglobin concentration
and the degree of ST change. He postulated that the
effect may have been mediated by an increase in
10.8 Treatment studies in men with coronary artery blood flow consequent upon vaso-
atherosclerosis dilation. The dose of testosterone used in this study
was probably supraphysiological. Webb et al. (1999)
10.8.1 Angina pectoris used quantitative coronary angiography to show that
The observational studies and animal experiments increasing doses of intra-coronary infusions of testos-
described above indicate that men with a low level terone caused progressive coronary vasodilatation.
of circulating testosterone have an increased athero- Rosano et al. (1999) and Webb et al. (1999b) studied
genic profile, and demonstrably more atherosclerosis. the effect of intravenous testosterone on angina
The process of atherosclerosis in humans results in threshold using standard exercise testing, and showed
various clinical manifestations. First, sudden throm- that time to 1 mm ST segment depression was pro-
botic occlusion of arteries at the site of atherosclerotic longed after intravenous testosterone compared with
plaques causes acute coronary syndromes (myocar- both baseline and placebo.
dial infarction) and stroke. Epidemiological studies Our group showed for the first time in a random-
(described above) do suggest that these events are ized, placebo-controlled clinical trial that physio-
more common in men with lower levels of testoster- logical doses of testosterone improved angina
one, especially, for example, after prostate surgery. threshold but only significantly in those men with a
A study of men attending a urology clinic for ED low baseline level of testosterone (English et al.
(Corona et al. 2010) showed that men with a low 2000c). In this study men were selected only if they
testosterone level were more likely to have fatal car- had a reproducibly positive treadmill exercise test.
diovascular events during a relatively short follow-up They were treated with low dose physiological
period of just over four years. These authors found no replacement therapy by skin patches delivering 5 mg
statistical relationship between testosterone levels and testosterone daily in a slow-release form. This dose is
major cardiovascular events. sufficient to raise testosterone levels to normal in 93%
Second, progressive atherosclerosis causes fixed of hypogonadal patients. Treatment did increase total
narrowing of arteries which results in reduced blood testosterone levels but not outside the normal range.
flow to vital organs, and tissue ischemia. The classical There was no measurable difference in hemoglobin
clinical manifestation of this is angina pectoris. levels with this formulation. Exercise duration and
In men with hypogonadism, does TRT improve time to 1 mm ST segment depression was prolonged
symptoms of angina? in those men taking testosterone, but the effect was
Testosterone therapy has been used in men with most marked in those men with low starting levels. In
angina since the 1940s, with observational evidence of a repeat study in men with hypogonadism and
improvement. In one study of 100 patients with angina, and using a crossover methodology, we found
angina (including eight women), Lesser (1946) much more marked improvements (Malkin et al.

217
Chapter 10: Testosterone and cardiovascular disease

Table 10.1 Review of all published studies of testosterone in men with angina

Author Drug dosage Primary outcome n Result


Hamm (1942) Variable 7 Decreased frequency of angina
Walker (1942) Variable 9 Increased exercise tolerance
Sigler (1943) Low 16 Increased exercise duration
Lesser (1946) Low 92 Improvement in 85
Jaffe (1977) Physiological Exercise test 50 Decreased ST depression
Wu and Weng (1993) Variable Holter 62 Decreased ischemia on Holter
Rosano et al. (1999) High Exercise test 14 Increased time to ischemia
Webb et al. (1999b) High Exercise test 14 Increased time to ischemia
English et al. (2000c) Physiological Exercise test 46 Increased time to ischemia
Thompson et al. (2002) High and physiological Exercise test SPECT scan 32 Neutral
Malkin et al. (2004a) Physiological Exercise test 10 Increased time to ischemia
Mathur et al. (2009) Physiological Exercise test 13 Increased time to ischemia
Abbreviations: SPECT, single photon emission computed tomography.

2004a) in all men despite concomitant anti-anginal suggests downregulation of the dilatory response after
therapy. All these clinical studies report improve- replacement therapy, which was not seen in the clin-
ments over the short-term (three months). The dur- ical study where the improvement in angina threshold
ability of the response was tested in a long-term study was persistent at 12 months. Laboratory studies indi-
of 12 months duration using depot injections every cate that testosterone causes vasodilatation by inter-
3 months (testosterone undecanoate Nebido) in action with the L-type calcium channel in the smooth
men with chronic stable angina and reproducibly low muscle cell membrane of vessels, which is the site of
early morning testosterone blood level (<12 nmol/l) action for the drug nifedipine, which is also a known
(Mathur et al. 2009). This randomized, placebo- anti-anginal agent (Scragg et al. 2004).
controlled, parallel-group study showed significant Additional clinical benefits that have been seen in
and persistent improvement in exercise duration and these men treated with replacement doses of testoster-
time to 1mm ST segment depression on treadmill one include favorable modifications to standard
testing with active treatment. accepted risk factors including:
In all there have been 12 studies of the use of  total cholesterol (see Chapter 11 for further
testosterone therapy in men with angina with consist- details) and LDL (Malkin et al. 2004b);
ently positive results. An overview of these trials has  obesity and insulin resistance (Malkin et al. 2007);
shown that the size of the clinical benefit is greater in  improved fibrinolysis (Smith et al. 2005);
those men with low baseline levels of testosterone
 reduced inflammation/cytokine activation
(<12 nmol/l), and the effect lasts for at least one year (Malkin et al. 2004b);
(see Table 10.1).
 improved diabetic control (Kapoor et al. 2006;
The mechanism underpinning the improvement
Jones et al. 2011).
in exercise duration and the direct anti-ischemic
effect seen in these clinical studies is probably coron-
ary artery vasodilatation. In vitro, isolated human 10.9 Testosterone and heart failure
arteries from hypogonadal men show an augmented Chronic heart failure (CHF) can best be described as a
vasodilatation to testosterone compared with control syndrome characterized by impairment of cardiac
arteries from eugonadal men (Malkin et al. 2006a). function associated with a maladaptive metabolic and
After three months of physiological replacement ther- neuro-hormonal axis. Chronic heart failure is a
apy this augmented response was less obvious. This common clinical problem and a major public health

218
Chapter 10: Testosterone and cardiovascular disease

Table 10.2 Metabolic changes in heart failure

Metabolic axis Specific compounds Relationship Clinical effect in Effect of Available


with chronic heart failure pharmacological therapies
heart failure modification
Renin- Angiotensin II Relates to Vasoconstriction, Improves ACE-I
angiotensin severity of fluid retention, symptoms and ARBs
CHF and myocardial reduces mortality
predicts fibrosis
deterioration
and mortality
Aldosterone Myocardial Improves Spironolactone
fibrosis symptoms and Eplerenone
reduces mortality
Catecholamines Adrenalin Relate to Increase risk of Improve b-Blockers
Noradrenalin severity of sudden death, symptoms and
CHF and worsen cardiac reduce mortality
predict function
deterioration,
mortality and
sudden death
Glucocorticoid Cortisol Elevated Catabolic Unknown None
Insulin Resistance to Impaired glucose Unknown
insulin action delivery,
in proportion catabolic
to severity of
CHF
Growth hormone Reduced in Catabolic Recombinant
heart failure, growth
resistance at hormone
receptor level
Androgens Testosterone Reduced in Catabolic Improves Testosterone
heart failure endurance
Dehydroepiandrosterone Reduced in Catabolic Unknown
heart failure
Immune/ Tumor necrosis factor Elevated Catabolic None Monoclonal
cytokine antibodies
Interleukin 1 Elevated Catabolic None Monoclonal
antibodies
Abbreviations: ACE-I, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; b-blocker, beta-adrenoceptor blocker.

issue. The prevalence of CHF in the UK is 1% and in of angiotensin-converting enzyme inhibitors, beta-
Europe alone it is thought that around 10 million adrenergic receptor blockers and direct aldosterone
people are affected by CHF (Swedberg et al. 2005), antagonists which improve symptoms and prolong life,
imposing a financial burden in the UK accounting but do not prevent eventual decompensation to pro-
for 45% of the National Health Service budget. In gressive heart failure. Other widely used therapies such
most patients the clinical condition is characterized as loop diuretics, digoxin and other anti-arrhythmics
by relentlessly progressive breathlessness, worsening may improve symptoms and keep patients from
exercise tolerance and overwhelming fatigue. Modern unwanted hospital admission but have no demon-
heart failure drug therapy uses combinations strable effect on mortality (Swedberg et al. 2005).

219
Chapter 10: Testosterone and cardiovascular disease

Despite modern advances in the detection, diag- catabolic hormones and a deficiency of many anabolic
nosis and treatment of CHF, the prognosis of this hormones, which is associated with a poorer progno-
condition is still poor and is no better than the prog- sis and significant morbidity (Anker et al. 1997).
nosis of most malignancies. Severe heart failure which Jankowska et al. (2006) showed that hormonal defi-
is characterized by breathlessness at rest or on min- ciencies in gonadal, adrenal and somatotropic hor-
imal exertion has an annualized mortality of 50% mones in men with heart failure were independent
(Swedberg et al. 2005). markers of poor prognosis, but it is not clear whether
these changes are the cause or the result of heart
10.9.1 Pathophysiology failure. Whether cause or effect, when testosterone
In order to appreciate the potential role for testosterone levels are low in men with heart failure then this is a
in the treatment of men with heart failure, it is neces- poor prognostic marker (Guder et al. 2010; Wehr
sary to understand some background pathophysiology et al. 2011). About a quarter to a third of men with
of the condition. Chronic heart failure is a unique moderate-severity heart failure have biochemical evi-
metabolic syndrome characterized by perturbation of dence of testosterone deficiency (Kontoleon et al.
numerous endocrine and inflammatory parameters. 2003; Malkin et al. 2006b). One of the most important
These changes are important since they relate to the features of severe heart failure is a severe anabolic
severity of heart failure and directly contribute to catabolic imbalance resulting in cardiac cachexia
deterioration and prognosis. A summary of the heart (Anker et al. 1997), which is defined clinically as the
failure metabolic syndrome is displayed in Table 10.2. non-intentional loss of 6 kg lean mass over a six-
The physiological consequence of inadequate car- month period. Cachexia is the most extreme symp-
diac pumping action is reduced cardiac output, tom of heart failure, and most subjects experience a
resulting in low organ perfusion pressure. This results more gradual catabolic decline. Cardiac cachexia is
in neuro-humeral activation of systems causing fluid strongly associated with testosterone deficiency. The
retention and vasoconstriction, which aim to return the clinical characteristics of muscular wasting and weak-
cardiac output and blood pressure to normal. However, ness in heart failure show that the condition is not
many of these adaptive changes are ultimately detri- simply a disease of the heart: there are multi-system
mental and cause worsening of cardiac function and effects. Many of the cardinal symptoms of heart fail-
eventually lead to deterioration in the severity of heart ure such as breathlessness and fatigue are due to
failure. Strategies to simply increase cardiac muscle abnormal muscle function, impaired mobilization of
contraction with inotropes, for example merely energy and ultimately loss of lean muscle mass (Coats
accelerate the decline in function leading to a worse 2001). It is these features of heart failure, intuitively
outcome. The mainstay of modern drug therapy is to similar to a state of frank androgen deficiency, which
interfere pharmacologically with these adaptive changes led to the idea of using testosterone treatment as a
and so to delay the inevitable cardiac decline. Symptoms catabolic antagonist.
are controlled and deterioration slowed by inhibition of
the renin-angiotensin-aldosterone axis and blockade of
catecholamine receptors, as summarized in Table 10.2. 10.9.3 Theoreticalbasisfor testosteroneas
Disordered and excess immune activation has been a treatment for heart failure
explored with small trials of immunoglobulin and also
Testosterone is a logical choice as a treatment for
pentoxifylline, with varying benefit. The possibility of
heart failure, as the condition is characterized by
reducing pro-inflammatory cytokines has been inten-
anabolic deficiency, low-grade inflammation and a
sively investigated, and although initial studies of
loss of muscle mass and strength. These are effects
anti-TNF biological drugs were promising, defini-
that testosterone treatment, even at physiological
tive benefit has not been borne out in major clinical
doses, may improve. In addition, in heart failure there
trials (Anker and Coats 2002).
is systemic peripheral vasoconstriction resulting in
increased systemic vascular resistance. As discussed
10.9.2 Androgen status in heart failure above, since testosterone is also a direct coronary and
Patients with heart failure have been shown to have a peripheral vasodilator, this action should improve
clear anaboliccatabolic imbalance, with an excess of cardiac output and function.

220
Chapter 10: Testosterone and cardiovascular disease

10.9.4 Cardiac effects of testosterone In one recent interventional study of frail hypo-
gonadal men, supraphysiological dosages of TRT
treatment were used in an attempt to improve muscle strength
Within the boundaries of the normal physiological (Basaria et al. 2010). The study was positive in show-
range, testosterone therapy seems to have relatively ing significant increases in muscle strength but was
little effect on myocardial morphology and function. stopped early because of an excess of cardiovascular
There are animal data that profound testosterone side-effects. The authors reported that 23 patients
depletion (castration) results in reduced left ventricu- taking testosterone had cardiovascular complications
lar mass, reduced cardiac output and reduced ejection compared with 5 in the placebo group, and on this
fraction (Scheuer et al. 1987), possibly through the basis stopped the trial. Critical review of this paper
expression of certain calcium ion channels and shows that in fact there were only 6 hard end-points
protein synthesis. In Klinefelter syndrome, echocar- in the treatment group compared with 1 in the pla-
diographic data indicate that systolic function is cebo group. About half of the group had a history of
impaired and systolic velocities correlate with testos- cardiovascular disease, and the rest had significant
terone levels (Andersen et al. 2008). There are consid- cardiovascular risk factors. If this study shows any-
erably more data on the effects of supraphysiological thing it is that men with hypogonadism should be
testosterone treatment on the myocardium. There is treated only with physiological doses of testosterone
general uniformity in cell culture studies, intact for true replacement therapy. The literature shows
animal treatment studies and observational studies that testosterone replacement should be managed in
of power athletes known to abuse anabolic androgens, the same way that thyroid hormone replacement is.
showing that a very high dose damages the myocar- Replacement dosages should aim to maintain normal
dium. The important specific findings from the physiological levels. If the Basaria trial had been
human observational studies include increased left conducted in hypothyroid patients with high cardio-
ventricular mass and hypertrophy, smaller ventricular vascular risk and replacement had aimed at supraphy-
cavity dimensions and evidence of early diastolic dys- siological levels, the same (or worse) result would
function due to stiffening and loss of left ventricular have been seen.
compliance (Urhausen et al. 2004). In patients with heart failure, skeletal muscle is
abnormal with muscle fiber atrophy and a shift from
10.9.5 Non-cardiac effects of testosterone oxidative to glycolytic metabolism. Some authorities
believe that these muscle changes result in autonomic
treatment and ventilatory responses which contribute to symp-
There have been numerous clinical trials of androgen toms of breathlessness and fatigue. These changes can
therapy on the effects of body composition and vol- be temporarily reversed by exercise training. At the
untary physical strength. The trials can be broadly cellular level, a potential site of action for testosterone
divided into those using physiological or non-physio- treatment has been identified which involves the local
logical testosterone therapy and those testing the expression of IGF-1 and the nuclear accumulation
effects in morbid populations, androgen-deficient of pro-myogenic, anti-adipogenic stem cell regulator,
males and normal subjects. There is a general consist- b-catenin (Gentile et al. 2010). These findings
ency within the literature, with well-conducted, pro- strongly suggest that the anabolic effect of physio-
spective randomized controlled trials showing that logical doses of testosterone is likely to be beneficial
testosterone improves anabolic function. This for patients with heart failure.
improvement in function is characterized by
increased voluntary muscle strength, increased lean
(muscle) mass and reduced fat mass (Isidori et al. 10.9.6 Insulin resistance
2005). These effects are seen in all patient groups One of the major hormonal derangements in heart
and with testosterone preparations within the physio- failure is resistance to the action of insulin, which is
logical replacement range. The morbid populations another example of a maladaptive humeral change in
studied include patients with weight loss and cachexia the complex syndrome of heart failure. The severity
due to malignancy, HIV infection and inflammatory of heart failure is related to the severity of the insulin
autoimmune disease. resistance, and impaired insulin-mediated glucose

221
Chapter 10: Testosterone and cardiovascular disease

uptake is a powerful independent prognostic marker invasive hemodynamic monitoring in 12 patients with
(Anker et al. 1997). The mechanism underlying insu- chronic heart failure randomized to six hours of acute
lin resistance in heart failure is obscure, though it testosterone therapy or placebo in random order, has
appears functionally different from insulin resistance confirmed the thesis that testosterone reduces sys-
in other morbid populations, which in general are temic vascular resistance and consequently increases
characterized by reduced phosphorylation of intra- cardiac index (Fig. 10.2) (Pugh et al. 2003). The levels
cellular post-insulin receptor proteins. It is likely that of testosterone in the treatment phase of this study
the cause is multifactorial and involves impaired were in the high physiological range and show that, in
post-receptor signaling and other neuro-hormonal the short term at least, testosterone increases cardiac
and immune alterations present in severe heart fail- output as a result of reducing vascular resistance and
ure (Kemppainen et al. 2003). It is notable that increasing myocardial stroke volume. Further analysis
insulin resistance can be improved by both conven- of these data demonstrates that the patients with the
tional heart failure treatments such as angiotensin lowest baseline testosterone levels (patients below the
converting enzyme inhibitors and b-adrenergic median) derived a greater hemodynamic effect. Pul-
receptor blockers as well as non-pharmacological monary vascular resistance is frequently elevated in
treatments such as graded exercise. Testosterone men with chronic left ventricular failure, due in part to
treatment has a positive effect on insulin sensitivity dysregulation of smooth muscle tone. There is in vitro
both in normal subjects and morbid populations evidence in human pulmonary arteries and veins that
such as obese men and diabetics (Kapoor et al. physiological levels of testosterone act as a vasodilator.
2005; Jones et al. 2011). In a small placebo-controlled Potentially this effect of testosterone could contribute
crossover study in men with chronic heart failure to the beneficial effect of testosterone observed in
(see Chapter 11), testosterone improved fasting clinical trials (Jones et al. 2002; Smith et al. 2008;
glucose and insulin levels, and insulin resistance Rowell et al. 2009).
as measured by HOMA (Homeostatic Model
Assessment), and this was associated with increased 10.9.9 Erythropoietic effects of
lean mass and reduced fat mass (Malkin et al. 2007).
testosterone
10.9.7 Inflammation As discussed above, one of the consequences of
chronic heart failure is persistent chronic anemia. This
There is low-grade, subclinical inflammatory acti-
is usually mild, but in some patients (particularly those
vation in chronic heart failure. Inflammatory medi-
with cardiac cachexia) it can be an intractable problem.
ators such as tumor necrosis factor (TNF-a) and
Indeed, the problem of anemia has prompted studies
interleukins such as IL-1 and IL-6 are elevated in
of the role of erythropoietin in patients with heart
patients with heart failure, and contribute to cachexia
failure. A meta-analysis of the available randomized,
and insulin resistance (Anker et al. 1997). Testoster-
controlled trial evidence does show a small benefit
one replacement therapy reduces blood levels of
when erythropoietin is given to patients with heart
inflammatory cytokines in hypogonadal men with
failure, compared with placebo (Kotecha et al. 2011).
co-morbid disease such as diabetes and coronary dis-
Testosterone increases hemoglobin in a dose-respon-
ease (Malkin et al. 2004b). However, significant clin-
sive way, and this effect is beneficial in these patients.
ical effects in men with heart failure have not been
In the treatment trials in heart failure (Malkin
detected in vivo (Pugh et al. 2005), although clinical
et al. 2006b; Caminiti et al. 2009), men given TRT
trials at present are limited and under-powered.
had a small but statistically significant increase in
hematocrit by about 1.5% over placebo, but no
10.9.8 Hemodynamics in heart failure change in hemoglobin level.
Testosterone therapy has beneficial effects on the
hemodynamics of heart failure. Experimental human 10.9.10 Clinical trials of testosterone
data have confirmed that testosterone in vitro is a
dilator of pre-constricted systemic vessels (isolated therapy in heart failure
from subcutaneous fat) (Malkin et al. 2006a). Further- There are few trials of androgen treatment in heart
more, an important in-vivo crossover study, using failure. A single animal study using a low dose of

222
Chapter 10: Testosterone and cardiovascular disease

(A) Fig. 10.2 Figure showing the acute


hemodynamic changes seen on
Treatment effect (% change in cardiac index)

35 (A) cardiac index and (B) systemic vascular


resistance (SVR) after high-dose buccal
30 testosterone in men with heart failure.
Above median The data show the effect of testosterone
Below median in men with baseline bioavailable
25
p < 0.0001 testosterone above or below the median
of 4.6 nmol/l. From Pugh et al. (2003).
20

15

10

5
0 30 60 120 180 240 300 360
Time (minutes)

(B)
20

10
Treatment effect (% change in SVR)

10

20

30
Above median
40 Below median

p < 0.0001
50

60
0 30 60 120 180 240 300 360
Time (minutes)

nandrolone decanoate improved survival in male improve mood, symptom scores and endurance
hamsters with an inherited cardiomyopathy. An (using an incremental shuttle walk test). In the larger
unblinded descriptive study of 12 male patients found follow-up study to this pilot (Malkin et al. 2006b),
improvements in echocardiographic parameters using low-dose daily transdermal testosterone patches
(reduced left ventricular diameter, reduced left ven- (Androderm 5 mg or placebo), the effect on endur-
tricular mass) and reductions in brain natriuretic ance was confirmed. Testosterone was found to
peptide (a serum marker of heart failure severity). increase exercise capacity as measured with an incre-
The first prospective, double-blind studies of tes- mental shuttle walk test. The improvement in exercise
tosterone treatment in heart failure were from the capacity in this study was less than observed in the
same scientific research group. In a pilot study using pilot, in which larger doses of testosterone were given,
injections of im testosterone (Sustanon 100) every suggesting that there may be a dose-response relation-
fortnight, Pugh et al. (2004) found testosterone to ship. Examination of the pooled data confirms that

223
Chapter 10: Testosterone and cardiovascular disease

the patients treated with im testosterone achieved Heart failure is a condition of high mortality,
higher serum levels of testosterone and greater chronic debilitating symptoms and recurrent hospi-
increases in functional capacity. These data suggest talization. Novel therapies should either improve
that the biological effects of testosterone on functional morbidity or survival or both; ideally therapy should
exercise capacity are related to the serum levels reached be widely applicable, inexpensive and show benefit in
in vivo. Furthermore, consistent with this notion, there the presence of coexisting heart-failure therapies.
was a positive correlation with the increase in exercise Although TRT cannot be advocated for women, tes-
capacity and serum bioavailable testosterone at three tosterone treatment is indicated in at least 25% of
and six months. Other secondary outcomes from this men with heart failure who will have a low testoster-
study included improved physician-assigned symp- one blood level. These men all have symptoms com-
tom scores, increased voluntary muscle strength, patible with hypogonadism, so there is no reason to
increased left ventricular cavity length by withhold therapy, once prostate malignancy had been
echocardiography, and a trend to a reduction in left excluded. In those men with a testosterone blood level
ventricular mass. Systolic blood pressure fell in the within the normal range it can be argued that, within
placebo group over the 12 months follow-up, which is the context of the hormonal imbalance of heart fail-
a recognized part of the natural history of heart failure ure, this represents a relative androgen deficiency.
and is an adverse prognostic sign; systolic blood pres- However, as discussed above it is important to only
sure in the testosterone group was maintained. use physiological doses of replacement therapy to
There have been two further studies of TRT in avoid potential complications.
patients with heart failure one in men and one in
women. In a short-term study of three-months dur- 10.10 Testosterone levels and
ation, Caminiti et al. (2009) examined the effects of
TRT or placebo using a long-acting depot testosterone mortality
in 70 men (average age 70 years) with heart failure in The disadvantage of having a low blood testosterone
association with reduced systolic function on echocar- level is not only related to the development of accel-
diography (<40% mean ejection fraction 32%). Only erated atherosclerosis. Ruige et al. (2011) have
30% were clinically and biochemically hypogonadal. reviewed the published literature on overall mortality
They used cardio-pulmonary exercise test and six- and testosterone level up to 2009. There does appear
minute walk tests to assess functional improvement, to be an excess mortality in elderly men (>70 years of
which improved in those men taking TRT, in relation age) who have a low testosterone level, which is espe-
to the change in testosterone blood level. cially marked in studies published since 2007. Five
They examined the relationship between oxygen recent studies have demonstrated that lower baseline
consumption and ventilation which is deranged in testosterone levels are a significant predictive marker
men with heart failure, and showed that TRT reduced for mortality even after controlling for the effects of
the gradient of the ventilation/CO2 elimination rate co-morbid conditions. In 2004, Shores et al. (2004)
slope, which implies a beneficial effect of testosterone reported that hypotestosteronemia was a marker for
on ventilatory efficiency. Effectively this means that men mortality in a group of 44 geriatric inpatients within a
treated with TRT felt less breathless per unit of exercise six-month period. In a follow-on study, the same
compared with their peers on placebo. These authors group performed a computerized analysis of the Vet-
also showed that TRT improved baroreceptor sensitiv- erans Affairs clinical database (Shores et al. 2006),
ity, which is a novel finding in humans. As baroreceptor including 850 men followed up over a four- to eight-
sensitivity is decreased in heart failure (and associated year period. After controlling for co-morbid condi-
with an adverse prognosis), such a change has theoretical tions which would affect mortality, e.g. concurrent
advantages. The greatest benefits were seen in men with cancer, they found that men with low testosterone
lower baseline levels of testosterone. levels had an 88% (20.1 vs. 34.9%, p < 0.001) relative
This research group has also used low-level TRT increase in all-cause mortality risk when compared
in a six-month study of women with heart failure and with those with normal testosterone levels at baseline.
showed similar effects. The safety of testosterone ther- In 2007, the InCHIANTI study demonstrated that an
apy in women with heart failure in the long term has age-associated fall in bioavailable testosterone was
not been assessed (Iellamo et al. 2010). associated with increased risk of death (Maggio et al.

224
Chapter 10: Testosterone and cardiovascular disease

(A) Fig. 10.3 Survival curves for mortality


Survival by testosterone status. Bio-T/all-cause mortality (adjusted) based on baseline bioavailable
testosterone (bio-T). (A) All-cause
1.00 mortality; (B) vascular mortality. The solid
line represents patients with baseline
bio-T less than 2.6 nmol/l; the broken line
represents patients with bio-T greater
0.95 than 2.6 nmol/l. HR, hazard ratio (From
Bio-T > 2.6 nmol/l Malkin et al. 2010).

0.90
Cumulative survival

0.85

Bio-T < 2.6 nmol/l


0.80

0.75

0.70 log rank, p < 0.0001, HR 2.2 (1.4 3.6)

0 500 1000 1500 2000 2500 3000 3500


Survival time (days)

(B)
Survival by testosterone status. Bio-T/vascular mortality (adjusted)

1.000

0.975

Bio-T > 2.6 nmol/l


Cumulative survival

0.950

0.925

Bio-T < 2.6 nmol/l


0.900

0.875

log rank, p = 0.007, HR 2.2 (1.2 3.9)

0 500 1000 1500 2000 2500 3000 3500


Survival time (days)

225
Chapter 10: Testosterone and cardiovascular disease

2007). In a six-year follow-up study of 410 men aged vascular disease. However, in the recently published
over 65 years, they found that this effect was made study by Malkin et al. (2010), 930 men with angiogra-
more pronounced and more statistically significant phically proven CAD were prospectively followed up
when low testosterone was associated with similar over a seven-year period. These authors observed base-
declines in IGF and DHEAS. In contrast to men with line prevalence of hypogonadism in this group (by a
all three hormones above the lowest quartiles, men strict criterion) to be 24%. In this androgen-deficient
with one, two or three hormones in the lowest quar- group the mortality was 21% versus only 12% in the
tiles were increasingly at more risk of death. eugonadal group (p = 0.002). Low bioavailable testos-
In 2008, Laughlin et al. studied an older group terone but not total testosterone significantly influ-
(mean age of 71 years) of 794 men over a period of up enced the all-cause and cardiovascular mortality after
to 20 years (Laughlin et al. 2008). They found a the multivariate analysis (Fig. 10.3), suggesting that this
significant fall in bioavailable testosterone but not is the more sensitive assay in detecting pathological
total testosterone with age. The risk of death was deficiency and risk. Low testosterone, therefore,
greater for men in the lowest baseline quartile of both appears to be a marker for increased mortality.
total and bioavailable testosterone compared with The effect of TRT on survival in men with or
those in the highest quartile. After adjusting for age, without cardiovascular disease is unknown, as clinical
adiposity, and lifestyle choices, the risk of death was trials have not been sufficiently powered or protracted
44% greater for the lowest compared to the highest to detect any change. Nevertheless, the current evi-
quartile of total testosterone (hazard ratio (HR): 1.44; dence base on physiological TRT in some morbid
95% CI: 1.121.84) and 50% higher for the lowest populations is compelling. Hypogonadal men with
compared to the highest quartile of bioavailable tes- type 2 diabetes, angina pectoris and heart failure have
tosterone (HR: 1.50; CI: 1.151.96). been shown to derive benefit. Given the costs of large,
In the largest study to date investigating the effects long-term randomized controlled trials, a registry of
of endogenous testosterone levels and mortality, the treated and untreated patients with any or all of these
European Prospective Investigation into Cancer conditions in association with biochemical testoster-
Norfolk study (EPIC-Norfolk) (Khaw et al. 2007) pro- one deficiency could provide useful data.
spectively investigated all-cause and cardiovascular
mortality in 11 606 healthy men between the ages of 10.11 Key messages
40 and 79 years at baseline. Over a 610-year follow-up  Men have a higher cardiovascular risk than women.
period, they observed a statistically significant associ-  Low testosterone is associated with accelerated
ation between baseline serum testosterone level and all- atherosclerosis in both animal and human studies.
cause (HR: 0.75; CI: 0.551.00), cardiovascular (HR:  Low testosterone is associated with an adverse
0.62; CI: 0.450.84) and cancer-related (HR: 0.59; CI: atherogenic risk profile.
0.420.85) deaths (p < 0.001) for each association after  Replacement testosterone therapy to normal
controlling for co-morbid conditions and behaviors. physiological levels improves symptoms of
angina and heart failure in men with
They found that a 1 SD increase in baseline testosterone
hypogonadism.
( 6 nmol/l) was associated with an approximate 14%
 Low testosterone is a marker of increased
risk reduction in mortality over the study period. cardiovascular and all-cause mortality.
In 2010, two more long-term epidemiological stud-  Large, randomized controlled trials are needed
ies showing that a low serum testosterone is associated to test the hypothesis that physiological
with an excess mortality were published (Haring et al. testosterone replacement will reduce this
2010; Malkin et al. 2010). Most of these studies have adverse effect on mortality.
been in populations of normal men without overt

10.12 References Akishita M, Hashimoto M, Ohike Y,


Ogawa S, Iijima K, Eto M, Ouchi Y
Alexandersen P, Haarbo J, Byrjalsen I,
Lawaetz H, Christiansen C (1999)
Agledahl I, Hansen JB, Svartberg J (2010) Low testosterone level as a Natural androgens inhibit male
(2008) Postprandial triglyceride predictor of cardiovascular events in atherosclerosis: a study in castrated,
metabolism in elderly men with Japanese men with coronary risk cholesterol-fed rabbits. Circ
subnormal testosterone levels. Asian factors. Atherosclerosis 210:232236 Research 84:813819
J Androl 10:542549

226
Chapter 10: Testosterone and cardiovascular disease

Andersen NH, Bojesen A, Kristensen healthy men. J Clin Endocrinol long-term androgen suppression for
K, Birkebaek NH, Fedder J, Bennett Metab 79:561567 prostate cancer with high metastatic
P, Christiansen JS, Gravholt CH Barrett-Connor E (1992) Lower risk: 10-year results of an EORTC
(2008) Left ventricular dysfunction endogenous androgen levels and randomised study. Lancet Oncol
in Klinefelter syndrome is dyslipidemia in men with non- 11:10661073
associated to insulin resistance, insulin-dependent diabetes mellitus. Bourghardt J, Wilhelmson AS,
abdominal adiposity and Ann Int Med 117:807811 Alexanderson C, De Gendt K,
hypogonadism. Clin Endocrinol Verhoeven G, Krettek A, Ohlsson C,
(Oxf) 69:785e91 Barrett-Connor E, Khaw KT (1988)
Endogenous sex hormones and Tivesten A (2010) Androgen
Anderson FH, Francis RM, Faulkner K cardiovascular disease in men. receptor-dependent and
(1996) Androgen supplementation A prospective population-based independent atheroprotection by
in eugonadal men with study. Circulation 78:539545 testosterone in male mice.
osteoporosis effects of 6 months Endocrinology 151:54285437
of treatment on bone mineral Basaria S, Coviello AD, Travison TG,
Storer TW, Farwell WR, Jette AM, Bowles DK, Maddali KK, Ganjam VK,
density and cardiovascular risk Rubin LJ, Tharp DL, Turk JR,
factors. Bone 18:171177 Eder R, Tennstedt S, Ulloor J,
Zhang A, Choong K, Lakshman Heaps CL (2004) Endogenous
Anderson RA, Ludlam CA, Wu FCW KM, Mazer NA, Miciek R, Krasnoff testosterone increases L-type Ca2
(1995b) Haemostatic effects of J, Elmi A, Knapp PE, Brooks B, channel expression in porcine
supraphysiological levels of Appleman E, Aggarwal S, Bhasin G, coronary smooth muscle. Am J
testosterone in normal men. Hede-Brierley L, Bhatia A, Collins Physiol 287:H2091H2098
Thromb Haemostasis 74:693697 L, LeBrasseur N, Fiore LD, Bhasin S Bruck B, Brehme U, Gugel N, Hanke S,
Anderson TJ, Uehata A, Gerhard MD, (2010) Adverse events associated Finking G, Lutz C, Benda N,
Meredith IT, Knab S, De La Grange with testosterone administration. Schmahl FW, Haasis R, Hanke H
D, Lieberman EH, Ganz P, Creager N Engl J Med 363:109122 (1997) Gender-specific differences
MA, Yeung AC, Selwyn AP (1995a) in the effects of testosterone and
Bavenholm P, de Faire U, Landou C,
Close relation of endothelial estrogen on the development of
Efendic S, Nilsson J, Wilman B,
function in the human coronary atherosclerosis in rabbits.
Hamsten A (1998) A progression of
Arterioscler Thromb Vasc Biol
and peripheral circulations. J Am coronary artery disease in young
Coll Cardiol 26:12351241 17:21922199
male post-infarction patients is
Anker SD, Coats AJ (2002) How to linked to disturbances of Caminiti G, Volterrani, M, Iellamo F,
carbohydrate and lipoprotein Marazzi G, Massaro R, Miceli M,
RECOVER from RENAISSANCE?
The significance of the results of metabolism and to impaired Mammi C, Piepoli M, Fini M,
fibrinolytic function. Eur Heart J Rosano GMC (2009) Effect of long-
RECOVER, RENAISSANCE,
RENEWAL and ATTACH. Int 19:402410 acting testosterone treatment on
J Cardiol 86:123130 Bebo BF, Schuster JC, Vandenbark AA, functional exercise capacity, skeletal
Offner H (1999) Androgens alter muscle performance, insulin
Anker SD, Chua TP, Ponikowski P, resistance, and baroreflex sensitivity
Harrington D, Swan JW, Kox WJ, the cytokine profile and reduce
encephalitogenicity of myelin- in elderly patients with chronic
Poole-Wilson PA, Coats AJ (1997) heart failure. A double-blind,
Hormonal changes and catabolic/ reactive T cells. J Immunol
162:3540 placebo-controlled, randomized
anabolic imbalance in chronic heart study. J Am Coll Cardiol 54:919927
failure and their importance for Beer NA, Jakubowicz DJ, Matt DW,
cardiac cachexia. Circulation Beer RM, Nestler JE (1996) Caron P, Bennet A, Camare R, Louvet
96:526534 Dehydroepiandrosterone reduces JP, Boneu B, Sie P (1989)
plasma plasminogen activator Plasminogen activator inhibitor in
Arad Y, Badimon JJ, Badimon L, plasma is related to testosterone in
Hembree WC, Ginsberg HN (1989) inhibitor type 1 and tissue
plasminogen activator antigen in men. Metab Clin Exper
Dehydroepiandrosterone feeding 38:10101015
prevents aortic fatty streak men. Am J Med Sci 311:205210
formation and cholesterol Bolla M, Van Tienhoven G, Warde P, Chou TM, Sudhir K, Hutchison SJ,
accumulation in the cholesterol-fed Dubois JB, Mirimanoff RO, Storme Ko E, Amidon TM, Collins P,
rabbit. Arteriosclerosis 9:159166 G, Bernier J, Kuten A, Sternberg C, Chatterjee K (1996) Testosterone
Billiet I, Torrecilla JL, Pfeffer R, induces dilation of canine
Bagatell CJ, Heiman JR, Matsumoto coronary conductance and
Cutajar CL, Van der Kwast T,
AM, Rivier JE, Bremner WJ (1994) resistance arteries in vivo.
Collette L (2010) External
Metabolic and behavioral effects of Circulation 94:26142619
irradiation with or without
high dose exogenous testosterone in

227
Chapter 10: Testosterone and cardiovascular disease

Chute CG, Baron JA, Plymate SR, Kiel Davoodi G, Amirezadegan A, accelerated coronary atherosclerosis
DP, Pavia AT, Lozner EC, OKeefe Borumand MA, Dehkori MR, with dehydroepiandrosterone in the
T, MacDonald GJ (1987) Sex Kazemisaeid A, Yaminisharif A heterotopic rabbit model of cardiac
hormones and coronary artery (2007) The relationship between transplantation. Circulation
disease. Am J Med 83:853859 level of androgenic hormones and 87:261269
Coats AJ (2001) Heart failure: what coronary artery disease in men. English KM, Mandour O, Steeds RP,
causes the symptoms of heart Cardiovasc J Africa 18:362366 Diver MJ, Jones TH, Channer KS
failure? Heart 86:574578 Debing E, Peeters E, Duquet W, Poppe (2000a) Men with coronary artery
Corona G, Monami M, Boddi V, K, Velkeniers B, Van Den Brande P disease have lower levels of
Cameron-Smith M, Fisher A, de (2008) Men with atherosclerotic androgens than men with normal
Vita G, Melani C, Balzi D, Sforza A, stenosis of the carotid artery have coronary angiograms. Eur Heart J
Forti G, Mannucci E, Maggi M lower testosterone levels compared 21:890894
(2010) Low testosterone is with controls. Int Angiol 27:135141 English KM, Jones RD, Jones TH,
associated with an increased risk of Deenadayalu VP, White RE, Stallone Morice AH, Channer KS (2000b)
MACE lethality in subjects with JN, Gao X, Garcia AJ (2001) Aging reduces the responsiveness of
erectile dysfunction. J Sex Med Testosterone relaxes coronary coronary arteries from male Wistar
7:15571564 arteries by opening the large- rats to the vasodilatory action of
Corrales JJ, Almeida M, Burgo R, conductance, calcium-activated testosterone. Clin Sci 99:7782
Mories MT, Miralles JM, Orfao A potassium channel. Am J Physiol English KM, Steeds RP, Jones TH,
(2006) Androgen-replacement Heart C 281:H1720H1727 Diver MJ, Channer KS (2000c)
therapy depresses the ex vivo Demirbag R, Yilmaz R, Ulucay A, Unlu Low-dose transdermal testosterone
production of inflammatory D (2005) The inverse relationship therapy improves angina threshold
cytokines by circulating antigen- between thoracic aortic intima in men with chronic stable angina: a
presenting cells in aging type-2 media thickness and testosterone randomized, double-blind,
diabetic men with partial androgen level. Endocr Res 31:335344 placebo-controlled study.
deficiency. J Endocrinol
De Pergola G, De Mitrio V, Sciaraffia Circulation 102:19061911
189:595604
M, Pannacciulli N, Minenna A, English KM, Jones RD, Jones TH,
Coviello AD, Kaplan B, Lakshman KM, Giorgino F, Petronelli M, Laudadio Morice AH, Channer KS (2001)
Chen T, Singh AB, Bhasin S (2008) E, Giorgino R (1997) Lower Gender differences in the
Effects of graded doses of androgenicity is associated with vasomotor effects of different
testosterone on erythropoiesis in higher plasma levels of pro- steroid hormones in rat pulmonary
healthy young and older men. J Clin thrombotic factors irrespective and coronary arteries. Horm Metab
Endocrinol Metab 93:914919 of age, obesity, body fat Res 33:645652
Crews JK, Khalil RA (1999a) distribution, and related metabolic English KM, Jones RD, Jones TH,
Antagonistic effects of 17 beta- parameters in men. Metabolism Morice AH, Channer KS (2002)
estradiol, progesterone, and 46:12871293 Testosterone acts as a coronary
testosterone on Ca2+ entry Ding AQ, Stallone JN (2001) vasodilator by a calcium
mechanisms of coronary Testosterone-induced relaxation of antagonistic action. J Endocrinol
vasoconstriction. Art Thromb Vasc rat aorta is androgen structure Invest 25:455458
19:10341040 specific and involves K+ channel Gentile MA, Nantermet PV (2010)
Crews JK, Khalil RA (1999b) Gender- activation. J Appl Physiol Androgen mediated improvement
specific inhibition of Ca2+ entry 91:27422750 of body composition and muscle
mechanisms of arterial Dobrzycki S, Serwatka W, Nadlewski S, function involves a novel early
vasoconstriction by sex hormones. Korecki J, Jackowski R, Paruk J, transcriptional program
Clin Exp Pharmacol & Physiol Ladny JR, Hirnle T (2003) An including IGF-1, MGF and
26:707715 assessment of correlations between induction of beta-catenin. J Mol
DAgostino P, Milano S, Barbera C, Di endogenous sex hormone levels and Endocrinol 44:55e73
Bella G, La Rosa M, Ferlazzo V, the extensiveness of coronary heart Gliczynski S, Ossowski M,
Farruggio R, Miceli DM, Miele M, disease and the ejection fraction of Slowinska-Srzednicka J,
Castagnetta L, Cillari E (1999) Sex the left ventricle in males. Med Brzezinska A, Zgliczynski W (1996)
hormones modulate inflammatory Invest 50:162169 Effect of testosterone replacement
mediators produced by Eich DM, Nestler JE, Johnson DE, therapy on lipids and lipoproteins
macrophages. Ann NY Acad Sci Dworkin GH, Ko D, Wechsler AS, in hypogonadal and elderly men.
876:426429 Hess ML (1993) Inhibition of Atherosclerosis 121:3543

228
Chapter 10: Testosterone and cardiovascular disease

Glueck CJ, Glueck HI, Stroop D, Speirs in the arterial vessel wall. Research Group. JAMA
J, Hamer T, Tracy T (1993) Circulation 103:13821385 280:605613
Endogenous testosterone, Haring B, Volzke H, Steveling A, Krebs Iellamo F, Volterrani M, Caminiti G,
fibrinolysis, and coronary heart A, Felix SB, Schofl C, Dorr M, Karam R, Massaro R, Fini M,
disease risk in hyperlipidemic men. Nauck M, Wallaschofski H (2010) Collins P, Rosano GMC (2010)
J Lab Clin Med 122:412420 Low serum testosterone levels are Testosterone therapy in women
Gordon GB, Bush DE, Weisman HF associated with increased risk of with chronic heart failure. A pilot
(1988) Reduction of atherosclerosis mortality in a population-based double-blind, randomized, placebo-
by administration of cohort of men aged 2079. Eur controlled study. J Am Coll Cardiol
dehydroepiandrosterone. A study Heart J 31:14911501 256:13101316
in hypercholesterolaemic Isidori AM, Giannetta E, Greco EA,
Hatakeyama H, Nishizawa M,
New Zealand White rabbits with Gianfrilli D, Bonifacio V, Isidori A,
Nakagawa A, Nakano S, Kigoshi T,
aortic intimal injury. J Clin Invest Lenzi A, Fabbri A (2005) Effects of
Uchida K (2002) Testosterone
82:712720 testosterone on body composition,
inhibits tumor necrosis factor-
Grady D, Herrington D, Bittner V, alpha-induced vascular cell bone metabolism and serum lipid
Blumenthal R, Davidson M, Hlatky adhesion molecule-1 expression in profile in middle-aged men: a meta-
M, Hsia J, Hulley S, Herd A, Khan S, human aortic endothelial cells. FEBS analysis. Clin Endocrinol (Oxf)
Newby LK, Waters D, Vittinghoff E, Letters 530:129132 63:280293
Wenger N; HERS Research Group Jaffe MD (1977) Effect of testosterone
Hauner H, Stangl K, Burger K, Busch
(2002) Cardiovascular disease cypionate on postexercise ST
U, Blmer H, Pfeiffer EF (1991) Sex
outcomes during 6.8 years of segment depression. Br Heart J
hormone concentrations in men
hormone therapy (HERS II). JAMA 39:12171222
with angiographically assessed
288:4957
coronary artery disease Jankowska EA, Biel B, Majda J,
Guder G, Fratz S, Bauersach SJ, Allolio relationship to obesity and body fat Szklarska A, Lopuszanska M,
B, Ertl G, Angermann CE, Stork S distribution. Klin Wsch 69:664668 Medras M, Anker SD, Banasiak W,
(2010) Low serum androgens and Poole-Wilson PA, Ponikowski P
Herman SM, Robinson JC, McCredie
mortality in heart failure. Heart (2006) Anabolic deficiency in men
RJ, Adams MR, Boyer MJ,
96:504509 with chronic heart failure:
Celermajer DS (1997) Androgen
Haffner SM, Mykkanen L, Valdez RA, deprivation is associated with prevalence and detrimental impact
Katz MS (1993) Relationship of sex enhanced endothelium-dependent on survival. Circulation 114:1829
hormones to lipids and lipoproteins dilatation in adult men. Arterioscler 1837
in non-diabetic men. J Clin Thromb Vasc Biol 17:20042009 Jones RD, English KM, Pugh PJ,
Endocrinol Metab 77:16101615 Morice AH, Jones TH, Channer KS
Honda H, Unemoto T, Kogo H (1999)
Hak AE, Witteman JC, de Jong FH, Different mechanisms for (2002) Pulmonary vasodilatory
Geerlings MI, Hofman A, Pols HA testosterone-induced relaxation of action of testosterone: evidence of a
(2002) Low levels of endogenous aorta between normotensive and calcium antagonistic action.
androgens increase the risk of spontaneously hypertensive rats. J Cardiovasc Pharmacol 39:814823
atherosclerosis in elderly men: the Hypertension 34:12321236 Jones RD, Pugh PJ, Hall J, Channer KS,
Rotterdam study. J Clin Endocrinol Jones TH (2003a) Altered
Metab 87:36323639 Howell SJ, Radford JA, Adams JE,
Smets EM, Warburton R, Shalet SM circulating hormone levels,
Hall J, Jones RD, Jones TH, Channer (2001) Randomized placebo- endothelial function and vascular
KS, Peers C (2006) Selective controlled trial of testosterone reactivity in the testicular feminised
inhibition of L-type Ca2 channels replacement in men with mild mouse. Eur J Endocrinol
in A7r5 cells by physiological levels Leydig cell insufficiency following 148:111120
of testosterone. Endocrinology cytotoxic chemotherapy. Clin Jones RD, Pugh PJ, Jones TH, Channer
147:26752680 Endocrinol 55:315324 KS (2003b) The vasodilatory action
Hamm L (1942) Testosterone Hulley S, Grady D, Bush T, Furberg C, of testosterone: a potassium channel
propionate in the treatment of Herrington D, Riggs B, Vittinghof E opening or a calcium antagonistic
angina pectoris. J Clin Endocrinol (1998) Randomized trial of estrogen action? Br J Pharm 138:733744
2:325328 plus progestin for secondary Jones RD, Ruban LN, Morton IE,
Hanke H, Lenz C, Spindler KD, prevention of coronary heart disease Roberts SA, English KM, Channer
Weidemann W (2001) Effect of in postmenopausal women. Heart KS, Jones TH (2003c) Testosterone
testosterone on plaque development and Estrogen/progestin inhibits the prostaglandin F-2-alpha
and androgen receptor expression Replacement Study (HERS) mediated increase in intracellular

229
Chapter 10: Testosterone and cardiovascular disease

calcium in A7r5 aortic smooth testosterone on brachial arterial elderly men. J Clin Endocrinol
muscle cells: evidence of an vasoreactivity in men with coronary Metab 87:15501554
antagonistic action upon artery disease. Am J Cardiol Kontoleon PE, Anastasiou-Nana MI,
store-operated calcium 89:862864 Papapetrou PD, Alexopoulos G,
channels. J Endocrinol Kapoor D, Malkin CJ, Channer KS, Ktenas V, Rapti AC, Tsagalou EP,
178:381393 Jones TH (2005) Androgens, insulin Nanas JN (2003) Hormonal
Jones RD, English KM, Jones TH, resistance and vascular disease in profile in patients with congestive
Channer KS (2004a) Testosterone- men. Clin Endocrinol (Oxf) 63:239 heart failure. Int J Cardiol
induced coronary vasodilatation 250 87:179183
occurs via a non-genomic Kapoor D, Goodwin E, Channer KS, Kotecha D, Ngo K, Walters JA,
mechanism: evidence of a direct Jones TH (2006) Testosterone Manzano L, Palazzuoli A, Flather
calcium antagonistic action. Clin Sci replacement therapy improves MD (2011) Erythropoietin as a
107:149158 insulin resistance, glycaemic treatment of anemia in heart failure:
Jones RD, Jones TH, Channer KS control, visceral adiposity and systematic review of randomized
(2004b) The influence of hypercholesterolaemia in trials. Am Heart J 161:822831.e2
testosterone upon vascular hypogonadal men with type 2 Larsen BA, Nordestgaard BG, Stender
reactivity. Eur J Endocrinol diabetes. Eur J Endocrinol 154:899 S, Kjeldsen K (1993) Effect of
151:2937 906 testosterone on atherogenesis in
Jones TH, Saad F (2009) The effects of Karakitsos D, Patrianakos AP, De cholesterol-fed rabbits with similar
testosterone on risk factors for, and Groot E, Boletis J, Karabinis A, plasma cholesterol levels.
mediators of, the atherosclerotic Kyriazis J, Samonis G, Parthenakis Atherosclerosis 99:7986
process. Atherosclerosis 207:308327 FI, Vardas PE, Daphnis E (2006) Laughlin GA, Barrett-Connor E,
Jones TH, Arver S, Behre H, Buvat J, Androgen deficiency and Bergstrom J (2008) Low serum
Meuleman E, Irribarren IM, endothelial dysfunction in men with testosterone and mortality in older
Morales A, Volterrani M, Yellowlees end-stage kidney disease receiving men. J Clin Endocrinol Metab
A, Howell JD, Channer KS (2011) maintenance hemodialysis. Am 93:6875
Testosterone replacement in J Nephrol 26:536543
Lesser MA (1946) Testosterone
hypogonadal men with type 2 Keating NL, OMalley AJ, Smith MR propionate therapy in one hundred
diabetes and/or metabolic (2006) Diabetes and cardiovascular cases of angina pectoris. J Clin
syndrome (the TIMES2 study). disease during androgen Endocrinol 6:549557
Diabetes Care 34:828837 deprivation therapy for prostate
cancer. J Clin Onc 24:44484456 Levine GN, DAmico AV, Berger P,
Kabakci G, Yildirir A, Can I, Unsal I, Clark PE, Eckel RH, Keating NL,
Erbas B (1999) Relationship Kemppainen J, Tsuchida H, Stolen K, Milani RV, Sagalowsky AI, Smith
between endogenous sex hormone Karlsson H, Bjornholm M, MR, Zakai N; American Heart
levels, lipoproteins and coronary Heinonen OJ, Nuutila P, Krook A, Association Council on Clinical
atherosclerosis in men undergoing Knuuti J, Zierath JR (2003) Insulin Cardiology and Council on
coronary angiography. Cardiology signalling and resistance in patients Epidemiology and Prevention, the
92:221225 with chronic heart failure. J Physiol American Cancer Society, and the
550 (Pt 1):305315 American Urological Association
Kanda N, Tsuchida T, Tamaki K (1996)
Testosterone inhibits Khaw KT, Dowsett M, Folkerd E, (2010) Androgen-deprivation
immunoglobulin production by Bingham S, Wareham N, Luben R, therapy in prostate cancer and
human peripheral blood Welch A, Day N (2007) Endogenous cardiovascular risk: a science
mononuclear cells. Clin Exper testosterone and mortality due to all advisory from the American Heart
Immunol 106:410415 causes, cardiovascular disease, and Association, American Cancer
cancer in men: European Society and American Urological
Kanda N, Tsuchida T, Tamaki K (1997)
prospective investigation into Association: endorsed by the
Testosterone suppresses anti-DNA
cancer in Norfolk (EPIC-Norfolk) American Society for Radiation
antibody production in peripheral
Prospective Population Study. Oncology. Circulation 121:833840
blood mononuclear cells from
Circulation 116:26942701 Li S, Li X, Li Y (2008) Regulation of
patients with systemic lupus
erythematosus. Arthritis Rheum Khosla S, Atkinson EJ, Dunstan CR, atherosclerotic plaque growth and
40:17031711 OFallon WM (2002) Effect of stability by testosterone and its
estrogen versus testosterone on receptor via influence of
Kang SM, Jang Y, Kim JY, Chung N,
circulating osteoprotegerin and inflammatory reaction. Vascul
Cho SY, Chae JS, Lee JH (2002)
other cytokine levels in normal Pharmacol 49:1418
Effect of oral administration of

230
Chapter 10: Testosterone and cardiovascular disease

Li Z, Danis V, Brooks P (1993) Effect of The effect of testosterone Minnemann T, Schubert M, Hbler D,
gonadal steroids on the production replacement on endogenous Gouni-Berthold I, Freude S,
of IL-1 and IL-6 by blood inflammatory cytokines and lipid Schumann C, Oettel M, Ernst M,
mononuclear cells in vitro. Clin Exp profiles in hypogonadal men. J Clin Mellinger U, Sommer F, Krone W,
Rheumatol 11:157162 Endocrinol Metab 89:33133318 Jockenhvel F (2007) A four-year
Liva SM, Voskuhl RR (2001) Malkin CJ, Jones RD, Jones TH, efficacy and safety study of the long-
Testosterone acts directly on Channer KS (2006a) Effect of acting parenteral testosterone
CD4(+) T lymphocytes to increase testosterone on ex vivo vascular undecanoate Aging Male
IL-10 production. J Immunol reactivity in man. Clin Sci (Lond) 10:155158
167:20602067 111:265274 Mukherjee TK, Dinh H, Chaudhuri G,
Lusis A (2000) Atherosclerosis. Nature Malkin CJ, Pugh PJ, West JN, van Beek Nathan L (2002) Testosterone
407:233241 EJ, Jones TH, Channer KS (2006b) attenuates expression of vascular
Testosterone therapy in men with cell adhesion molecule-1 by
Ly LP, Jimenez M, Zhuang TN, conversion to estradiol by
Celermajer DS, Conway AJ, moderate severity heart failure: a
double-blind randomized placebo aromatase in endothelial cells:
Handelsman DJ (2001) A double- implications in atherosclerosis. Natl
blind, placebo-controled, controlled trial. Eur Heart J
27:5764 Acad Sci USA 99:40554060
randomized clinical trial of
transdermal dihydrotestosterone gel Malkin CJ, Jones TH, Channer KS Muller M, van den Beld AW, Bots ML,
on muscular strength, mobility, and (2007) The effect of testosterone on Grobbee DE, Lamberts SW, van der
quality of life in older men with insulin sensitivity in men with heart Schouw YT (2004) Endogenous sex
partial androgen deficiency. J Clin failure. Eur J Heart Fail 9:4450 hormones and progression of
Endocrinol Metab 86:40784088 carotid atherosclerosis in elderly
Malkin CJ, Pugh PJ, Morris PD, Asif S, men. Circulation 109:20742079
Maggio M, Lauretani F, Ceda GP, Jones TH, Channer KS (2010) Low
Bandinelli S, Ling SM, Metter EJ, serum testosterone and increased Murphy JG, Khalil RA (1999)
Artoni A, Carassale L, Cazzato A, mortality in men with coronary Decreased [Ca(2+)](i) during
Ceresini G, Guralnik JM, Basaria S, heart disease. Heart 96:18211825 inhibition of coronary smooth
Valenti G, Ferrucci L (2007) muscle contraction by 17beta-
Mrin P, Holmng S, Gustafsson C,
Relationship between low levels of estradiol, progesterone, and
Jnsson L, Kvist H, Elander A, Eldh
anabolic hormones and 6-year testosterone. J Pharmacol Exp Ther
J, Sjstrm L, Holm G, Bjrntorp P
mortality in older men: the aging in 291:4452
(1993) Androgen treatment of
the Chianti area (InCHIANTI) study. abdominally obese men. Obes Res Nathan L, Shi WB, Dinh H, Mukherjee
Arch Intern Med 167:22492254 1:245251 TK, Wang XP, Lusis AJ, Chaudhuri
Makinen J, Jarvisalo MJ, Pollanen P, G (2001) Testosterone inhibits
Mathur A, Malkin C, Saeed B,
Perheentupa A, Irjala K, Koskenvuo early atherogenesis by conversion
Muthasamy S, Jones TH, Channer
M, Makinen J, Huhtaniemi I, to estradiol: critical role of
KS (2009) The long term effect of
Raitakari OT (2005) Increased aromatase. Natl Acad Sci USA
testosterone replacement therapy on
carotid atherosclerosis in 98:35893593
angina threshold and atheroma in
andropausal middle-aged men. patients with chronic stable angina. Nettleship JE, Jones TH, Channer KS,
J Am Coll Cardiol 45:16031608 Eur J Endocrinol 161:443449 Jones RD (2007a) Physiological
Mkinen JI, Perheentupa A, Irjala K, testosterone replacement therapy
McCrohon JA, Jessup W, Handelsman
Pllnen P, Mkinen J, Huhtaniemi attenuates fatty streak formation
DJ, Celermajer DS (1999) Androgen
I, Raitakari OT (2008) Endogenous and improves high-density
exposure increases human
testosterone and serum lipids in lipoprotein cholesterol in the Tfm
monocyte adhesion to vascular
middle-aged men. Atherosclerosis mouse: an effect that is independent
endothelium and endothelial cell
197:688693 expression of vascular cell adhesion
of the classic androgen receptor.
Circulation 116:24272434
Malkin CJ, Pugh PJ, Morris PD, Kerry molecule-1. Circulation
KE, Jones RD, Jones TH, Channer 99:23172322 Nettleship JE, Pugh PJ, Channer KS,
KS (2004a) Testosterone McRobb L, Handelsman DJ, Heather Jones T, Jones RD (2007b) Inverse
replacement in hypogonadal men relationship between serum levels of
AK (2009) Androgen-induced
with angina improves ischaemic progression of arterial calcification interleukin-1beta and testosterone in
threshold and quality of life. Heart men with stable coronary artery
in apolipoprotein E-null mice is
90:871876 uncoupled from plaque growth and disease. Horm Metab Res 39:366371
Malkin CJ, Pugh PJ, Jones RD, Kapoor lipid levels. Endocrinology Ong PJ, Patrizi G, Chong WC, Webb
D, Channer KS, Jones TH (2004b) 150:841848 CM, Hayward CS, Collins P (2000)

231
Chapter 10: Testosterone and cardiovascular disease

Testosterone enhances flow- Pugh PJ, Jones RD, West JN, Jones TH, Ruige JB, Mahmoud AM, De Bacquer
mediated brachial artery reactivity Channer KS (2004) Testosterone D, Kaufman JM (2011) Endogenous
in men with coronary artery disease. treatment for men with chronic testosterone and cardiovascular
Am J Cardiol 85:269272 heart failure. Heart 90:446447 disease in healthy men: a meta-
Perusquia M, Villalon CM (1999) Pugh PJ, Jones RD, Malkin CJ, Hall J, analysis. Heart 97:870875
Possible role of Ca2+ channels in the Nettleship JE, Kerry KE (2005) Saigal CS, Gore JL, Krupski TL, Hanley
vasodilating effect of 5 beta- Physiologic testosterone therapy has J, Schonlau M, Litwin MS (2007)
dihydrotestosterone in rat aorta. no effect on serum levels of tumour Androgen deprivation therapy
Eur J Pharmacol 371:169178 necrosis factor-alpha in men with increases cardiovascular morbidity
Perusquia M, Hernandez R, Morales chronic heart failure. Endocr Res in men with prostate cancer. Cancer
MA, Campos MG, Villalon CM 31:271283 110:14931500
(1996) Role of endothelium in the Qiu Y, Yanase T, Hu H, Tanaka T, Satariano WA, Ragland KE, Van Den
vasodilating effect of progestins and Nishi Y, Liu M, Sueishi K, Eeden SK (1998) Cause of death in
androgens on the rat thoracic aorta. Sawamura T, Nawata H (2010) men diagnosed with prostate
Gen Pharmacol 27:181185 Dihydrotestosterone suppresses carcinoma. Cancer 83:11801188
foam cell formation and Scheuer J, Malhotra A, Schaible TF,
Phillips GB, Pinkernell BH, Jing TY
attenuates atherosclerosis Capasso J (1987) Effects of
(1994) The association of
development. Endocrinology gonadectomy and hormonal
hypotestosteronemia with coronary
151:33073316 replacement on rat hearts. Circ Res
artery disease in men. Arterioscler
Thromb 14:701706 Rosano GM, Sheiban I, Massaro R, 61:1219
Pagnotta P, Marazzi G, Vitale C, Scragg JL, Jones RD, Channer KS,
Phillips GB, Jing TY, Laragh JH, Sealey
Mercuro G, Volterrani M, Aversa A, Jones TH, Peers C (2004)
JE (1995) Serum sex hormone levels
Fini M (2007) Low testosterone Testosterone is a potent inhibitor of
and renin-sodium profile in men
levels are associated with coronary L-type Ca2+ channels. Biochem
with hypertension. Am J Hypertens
artery disease in male patients Bioph Res Comm 318:503506
8:626629
with angina. Int J Impot Res
Phillips GB, Pinkernell BH, Jing TY 19:176182 Scragg JL, Dallas ML, Peers C (2007)
(2004) Are major risk factors for Molecular requirements for L-type
Rosano GMC, Leonardo F, Pagnotta P, Ca2 channel blockade by
myocardial infarction the major
Pelliccia F, Panina G, Cerquetani E, testosterone. Cell Calcium 42:1115
predictors of degree of coronary
della Monica PL, Bonfigli B, Volpe
artery disease in men? Metabolism Shahani S, Braga-Basaria M, Basaria S
M, Chierchia SL (1999) Acute anti-
53:324359 (2008) Androgen deprivation
ischemic effect of testosterone in
Pour HRN, Grobbee DE, Bots ML, men with coronary artery disease. therapy in prostate cancer and
Verhaar HJJ, Muller M, van der Circulation 99:16661670 metabolic risk for atherosclerosis.
Schouw YT (2007) Circulating sex J Clin Endocrinol Metab
Ross R (1999) Atherosclerosis: an 93:20422049
hormone levels and aortic stiffness inflammatory disease. N Engl J Med
in men. JAGS 55:621637 340:115126 Shores MM, Moceri VM, Gruenewald
Pugh PJ, Jones RD, Jones TH, Channer DA, Brodkin KI, Matsumoto AM,
Rossouw JE, Anderson GL, Prentice Kivlahan DR (2004) Low
KS (2002a) Intrinsic responses of rat RL, LaCroix AZ, Kooperberg C,
coronary arteries in vitro: influence testosterone is associated with
Stefanick ML, Jackson RD, decreased function and increased
of testosterone, calcium and Beresford SA, Howard BV, Johnson
effective transmural pressure. mortality risk: a preliminary study
KC, Kotchen JM, Ockene J; Writing of men in a geriatric rehabilitation
Endocrine 19:155162 Group for the Womens Health unit. J Am Geriatr Soc 52:20772081
Pugh PJ, Channer KS, Parry H, Initiative Investigators (2002) Risks
Downes T, Jones TH (2002b) Bio- and benefits of estrogen plus Shores MM, Matsumoto AM, Sloan
available testosterone levels fall progestin in healthy KL, Kivlahan DR (2006) Low serum
acutely following myocardial postmenopausal women: principal testosterone and mortality in male
infarction in men: association with results. JAMA 288:321333 veterans. Arch Intern Med
fibrinolytic factors. Endocrine Res 166:16601665
Rowell KO, Hall J, Pugh PJ, Jones TH,
28:161173 Channer KS, Jones RD (2009) Sigler LHTJ (1943) Treatment of
Mechanisms of agonist-induced angina pectoris by testosterone
Pugh PJ, Jones TH, Channer KS (2003)
constriction in isolated human propionate. NY J Med 43:14241428
Acute haemodynamic effects of
testosterone in men with chronic pulmonary arteries. Vasc Pharmacol Simon D, Preziosi P, Barrett-Connor E,
heart failure. Eur Heart J 24:909915 51:812 Roger M, Saint-Paul M, Nahoul K,

232
Chapter 10: Testosterone and cardiovascular disease

Papoz L (1992) The influence of sex hormone levels in men are not Huhtasaari F, Hallmans G (1998)
aging on plasma sex hormones in associated with risk of venous High plasminogen activator
men: the Telecom Study. Am thromboembolism: the Tromso inhibitor and tissue plasminogen
J Epidemiol 135:783791 study. Eur J Endocrinol 160:833838 activator levels in plasma precede a
Simon D, Charles MA, Nahoul K, Swedberg K, Cleland J, Dargie H, first myocardial infarction in both
Orssaud G, Kremski J, Hully V, Drexler H, Follath F, Komajda M, men and women: evidence for the
Joubert E, Papoz L, Eschwege E Tavazzi L, Smiseth OA, Gavazzi A, fibrinolytic system as an
(1997) Association between plasma Haverich A, Hoes A, Jaarsma T, independent primary risk factor.
total testosterone and cardiovascular Korewicki J, Lvy S, Linde C, Lopez- Circulation 98:22412247
risk factors in healthy adult men: the Sendon JL, Nieminen MS, Pirard L, Thompson PD, Cullinane EM, Sady SP,
Telecom Study. J Clin Endocrinol Remme WJ; Task Force for the Chenevert C, Saritelli AL, Sady MA,
Metab 82:682685 Diagnosis and Treatment of Herbert PN (1989) Contrasting
Smith A, English K, Malkin C, Jones R, Chronic Heart Failure of the effects of testosterone and
Jones T, Channer K (2005) European Society of Cardiology stanozolol on serum lipoprotein
Testosterone does not adversely (2005) Guidelines for the diagnosis levels. JAMA 261:11651168
affect fibrinogen or tissue and treatment of chronic heart Thompson PD, Ahlberg AW, Moyna
plasminogen activator (tPA) and failure: executive summary (update NM, Duncan B, Ferraro-Borgida M,
plasminogen activator inhibitor-1 2005): The Task Force for the White CM, McGill CC, Heller GV
(PAI-1) levels in 46 men with Diagnosis and Treatment of (2002) Effect of intravenous
chronic stable angina. Eur Chronic Heart Failure of the testosterone on myocardial
J Endocrinol 152:285291 European Society of Cardiology. ischaemia in men with coronary
Eur Heart J 26:11151140 artery disease. Am Heart J 143:249
Smith AM, Bennett RT, Jones TH,
Cowen ME, Channer KS, Jones RD Taylor LG, Canfield SE, Du XL (2009) 256
(2008) Characterisation of the Review of major adverse effects of Tivesten A, Mellstrm D, Jutberger H,
vasodilatory action of testosterone androgen-deprivation therapy in Fagerberg B, Lernfelt B, Orwoll E,
in the human pulmonary men with prostate cancer. Cancer Karlsson MK, Ljunggren O,
circulation. Vasc Health Risk 115:23882399 Ohlsson C (2007) Low serum
Management 4:14591466 Tedgui A, Mallat Z (2001) Anti- testosterone and high serum
Smith JC, Bennett S, Evans LM, inflammatory mechanisms in the estradiol associate with lower
Kynaston HG, Parmar M, Mason vascular wall. Circ Res 88:877887 extremity peripheral arterial disease
MD, Cockcroft JR, Scanlon MF, Tedeschi-Reiner E, Ivekovic R, Novak- in elderly men. The MrOS Study in
Davies JS (2001) The effects of Laus K, Reiner Z (2009) Sweden. J Am Coll Cardiol
induced hypogonadism on arterial Endogenous steroid sex hormones 50:10701076
stiffness, body composition and and atherosclerosis of retinal Tripathy D, Shah P, Lakshmy R, Reddy
metabolic parameters in males with arteries in men. Med Sci Monit KS (1998) Effect of testosterone
prostate cancer. J Clin Endocrinol 15:211216 replacement on whole body glucose
Metab 86:42614267 Tenover JS (1992) Effects of utilisation and other cardiovascular
Spinedi E, Suescun MO, Hadid R, testosterone in the aging male. J Clin risk factors in males with idiopathic
Daneva T, Gaillard RC (1992) Endocrinol Metab 75:10921098 hypogonadotrophic hypogonadism.
Effects of gonadectomy and sex Horm Metab Res 30:642645
Tep-areenan P, Kendall DA, Randall
hormone therapy on the endotoxin- MD (2002) Testosterone-induced Tsai HK, DAmico AV, Sadetsky N,
stimulated hypothalamo-pituitary- vasorelaxation in the rat mesenteric Chen M-H, Carroll PR (2007)
adrenal axis: evidence for a arterial bed is mediated Androgen deprivation therapy for
neuroendocrine-immunological predominantly via potassium localised prostate cancer and the
sexual dimorphism. Endocrinology channels. Br J Pharmacol risk of cardiovascular mortality.
131:24302436 135:735740 J Nat Cancer Inst 99:15161524
Svartberg J, von Muhlen D, Mathiesen Tharp DL, Masseau I, Ivey J, Ganjam Urhausen A, Albers T, Kindermann W
E, Joakimsen O, Bonan KH, VK, Bowles DK (2009) Endogenous (2004) Are the cardiac effects of
Stensland-Bugge E (2006) Low testosterone attenuates neointima anabolic steroid abuse in strength
testosterone levels are associated formation after moderate coronary athletes reversible? Heart 90:496
with carotid atherosclerosis in men. balloon injury in male swine. 501
J Intern Med 259:576582 Cardiovasc Res 82:152160 Uyanik BS, Ari Z, Gumus B, Yigitoglu
Svartberg J, Braekkan SK, Laughlin Thogerson AM, Jansson JH, Boman K, MR, Arslan T (1997) Beneficial
GA, Hansen JB (2009) Endogenous Nilsson TK, Weinehall L, effects of testosterone undecanoate

233
Chapter 10: Testosterone and cardiovascular disease

on the lipoprotein profiles in Webb CM, McNeill JG, Hayward CS, Yang XC, Jing TY, Resnick LM,
healthy elderly men. Jap Heart J de Zeigler D, Collins P (1999a) Phillips GB (1993) Relation of
38:7382 Effects of testosterone on coronary haemostatic risk factors to other
Vikan T, Johnsen SH, Schirmer H, vasomotor regulation in men with risk factors for coronary heart
Njolstad I, Svartberg J (2009) coronary heart disease. Circulation disease and to sex hormones
Endogenous testosterone and the 100:16901696 in men. Arterioscler Thromb
prospective association with carotid Webb CM, Adamson DL, de Zeigler D, 13:467471
atherosclerosis in men: the Tromso Collins P (1999b) Effect of acute Yesilova Z, Ozata M, Kocar IH, Turan
study. Eur J Epidemiol 24:289295 testosterone on myocardial M, Pekel A, Sengul A, Ozdemir IC
Walker TC (1942) The use of ischemia in men with coronary (2000) The effects of gonadotropin
testosterone propionate and artery disease. Am J Cardiol treatment on the immunological
estrogenic substance in the 83:437439 features of male patients with
treatment of essential hypertension, Wehr E, Pilz S, Boehm BO, Marz W, idiopathic hypogonadotropic
angina and peripheral vascular Grammer T, Obermayer-Pietsch B hypogonadism. J Clin Endocrinol
disease. J Clin Endoc 2:560568 (2011) Low free testosterone is Metab 85:6670
Wang C, Swerdloff RS, Iranmanesh A, associated with heart failure Yue P, Chatterjee K, Beale C, Poole-
Dobs A, Snyder PJ, Cunningham G, mortality in older men referred for Wilson PA, Collins P (1995)
Matsumoto AM, Weber T, Berman coronary angiography. Eur J Heart Testosterone relaxes rabbit
N; Testosterone Gel Study Group Fail 13:482488 coronary arteries and aorta.
(2000) Transdermal testosterone gel Whitsel EA, Boyko EJ, Matsumoto Circulation 91:11541160
improves sexual function, mood, AM, Anawalt BD (2001) Zhang X, Wang LY, Jiang TY, Zhang
muscle strength and body Intramuscular testosterone esters HP, Dou Y, Zhao JH, Zhao H,
composition parameters in and plasma lipids in hypogonadal Qiao ZD, Qiao JT (2002) Effects
hypogonadal men. J Clin Endocrinol men: a meta-analysis. Am J Med of testosterone and 17-beta-
Metab 85:28392853 111:261269 estradiol on TNF-alpha-induced
Wang C, Nieschlag E, Swerdloff E, WHO (1997) Acute myocardial E-selectin and VCAM-1
Behre HM, Hellstrom WJ, Gooren infarction and combined oral expression in endothelial
LJ, Kaufman JM, Legros J-J, contraceptives: results of an cells. Analysis of the
Lunenfeld B, Morales A, Morely JE, international multi-centre underlying receptor pathways. Life
Schulman C, Thompson IM, case-control study: WHO Sci 71:1529
Weldner W, Wu FCW (2008) collaborative study of Zitzmann M, Nieschlag E (2007)
Investigation, treatment and cardiovascular disease and steroid Androgen receptor gene CAG
monitoring of late-onset hormone contraception. Lancet repeat length and body mass index
hypogonadism in males. Consensus 349:12021209 modulate the safety of long-term
statement. Eur J Endocrinol Wu FC, von Eckardstein A (2003) intramuscular testosterone
159:507514 Androgens and coronary artery undecanoate therapy in
Wang C, Ilani N, Arver S, McLachlan disease. Endocr Rev 24:183217 hypogonadal men. J Clin Endocrinol
RI, Soulis T, Watkinson A (2011) Metab 92:38443853
Wu SZ, Weng XZ (1993) Therapeutic
Efficacy and safety of the 2% effects of an androgenic preparation Zitzmann M, Brune M, Nieschlag E
formulation of testosterone topical on myocardial ischemia and cardiac (2002) Vascular reactivity in
solution applied to the axillae in function in 62 elderly male coronary hypogonadal men is reduced by
androgen-deficient men. Clin heart disease patients. Chin Med J androgen substitution. J Clin
Endocrinol (Oxf) 75:836843. DOI 106:415418 Endocrinol Metab 87:50305037
10.1111/j.13652265.2011.04152.x

234
Chapter
Testosterone, obesity, diabetes and

11 the metabolic syndrome


T. Hugh Jones and Kevin S. Channer

11.1 Introduction 235 11.7 Effect of testosterone replacement on insulin


11.2 Association of low testosterone with sensitivity 241
obesity 236 11.7.1 Obesity 241
11.3 Metabolic syndrome and type 2 11.7.2 Metabolic syndrome and/or type 2
diabetes 236 diabetes 242
11.3.1 The metabolic syndrome 236 11.8 Effect of testosterone on components of the
11.3.2 Testosterone and the metabolic metabolic syndrome and type 2 diabetes 242
syndrome 237 11.8.1 Central adiposity and body
11.3.3 Testosterone and type 2 composition 242
diabetes 237 11.8.2 Hyperglycemia 242
11.3.4 Androgen deprivation therapy for 11.8.3 Dyslipidemia 244
prostate carcinoma 238 11.8.4 Hypertension 244
11.4 Interactions between the hypothalamic- 11.8.5 Inflammation 244
pituitary-testicular axis and adipose tissue 238 11.9 Cholesterol and lipoproteins 245
11.5 Role of the androgen receptor 11.10 Erectile dysfunction 245
polymorphism 240 11.11 Clinical implications 245
11.6 Mechanisms of testosterone action on 11.12 Key messages 245
insulin sensitivity/resistance 240 11.13 References 246

11.1 Introduction cardiovascular risk factors, especially those which com-


Many epidemiological studies have found a high preva- prise the key components of the metabolic syndrome.
lence of low testosterone levels in men with obesity, Insulin resistance is the common biochemical
metabolic syndrome and type 2 diabetes mellitus. abnormality between these conditions and is in itself a
A significant proportion of men with these conditions well-recognized cardiovascular risk factor. Understand-
have been shown to have classical symptoms of testoster- ing the link between testosterone and obesity and insulin
one deficiency, providing a diagnosis of hypogonadism. resistance is fundamental to determining its role in the
Obesity is a major risk factor for the development of the pathophysiology of these disorders. A question which is
metabolic syndrome and eventually type 2 diabetes. Both constantly asked is whether or not a low testosterone
metabolic syndrome and type 2 diabetes are associated level is merely a consequence of obesity, which is a state
with increased morbidity and mortality from cardiovas- of inflammation, and/or does it promote adiposity?
cular disease. Indeed, three-quarters of men with type 2 A similar query addresses the low testosterone state
diabetes die as a result of coronary heart disease or stroke. associated with cardiovascular disease, cancer, HIV,
There is an accumulating volume of published research chronic pulmonary disease and other inflammatory dis-
which links testosterone deficiency with important orders, in that it could be a biomarker of illness, but does

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

235
Chapter 11: Obesity, diabetes and the metabolic syndrome

IDF NCEP ATP III

Essential: Three factors from:


*Central obesity waist >94 cm
*Central obesity waist >102 cm
Plus two from: Fasting glucose >5.6 mmol/l
Blood pressure >130/85
Fasting glucose >5.6 mmol/l Fasting triglycerides >1.7 mmol/l
Blood pressure >130/85 HDL cholesterol <1.03 mmol/l
Fasting triglycerides >1.7 mmol/l
HDL cholesterol <1.03 mmol/l
Fig. 11.1 Definitions of the metabolic syndrome which are in common usage. *These two definitions have recently been amalgamated, with
agreement that three out of the five criteria are required for the diagnosis. As there are ethnic and population differences in the degree of elevated waist
circumference, it is important that this is taken into account for country-specific measurements. In addition, drug treatment of elevated triglycerides, blood
pressure and fasting glucose and reduced HDL-C, even if normalized for each criterion, are included (Alberti et al. 2009). See plate section for color version.

testosterone substitution have therapeutic benefits? inversely with the accumulation of visceral fat, but not
Evidence from recent clinical intervention studies does other fat depots, on CT scan after 7.5 years follow-up in a
support beneficial effects on certain cardiovascular risk group of 110 men (Tsai et al. 2000). The Quebec Family
end-points as well as relief of hypogonadal symptoms. Study found negative correlations of total testosterone
with waist circumference, body fat mass and visceral and
subcutaneous fat in a group of 130 men (Blouin et al.
11.2 Association of low testosterone 2005). The Tromso study of 1548 men aged 2485 years
with obesity showed that greater waist circumference was associated
Increased body fat is a well-known clinical feature of with low SHBG as well as testosterone (Svartberg et al.
hypogonadism which is clearly manifest in eunuchoi- 2004a). There are only a small number of studies that
dal body habitus in men with overt hypogonadism have investigated the effect of weight loss on testosterone
(Allan and McLachlan 2010; MacDonald et al. 2010). levels. One study found that a massive weight loss of
Several epidemiological studies have demonstrated an between 26 and 129 kg over a period of between 5 and 39
inverse relationship of testosterone levels with BMI months led to an increase in free and total testosterone
and central obesity (Seidell et al. 1990; Pasquali et al. and SHBG (Strain et al. 1988)
1991; Simon et al. 1992; Haffner et al. 1993; Couillard
et al. 2000; Svartberg et al. 2004a). The gradual 11.3 Metabolic syndrome and
decline of testosterone with age has been shown to type 2 diabetes
be associated with an increase in truncal obesity as
demonstrated by measurement of waist circumfer- 11.3.1 The metabolic syndrome
ence (Khaw and Barrett-Connor 1992). Furthermore, The metabolic syndrome is a constellation of cardiovas-
ADT in the treatment of prostate carcinoma leads to cular risk factors which together are strongly associated
an increase in waist circumference and body fat mass. with the future development of coronary heart disease,
Many studies have used waist circumference as a myocardial infarction, stroke and sudden cardiac death.
measure of central obesity that includes subcutaneous These risk factors include central obesity, impaired
as well as visceral fat. Central adiposity, however, does fasting glucose or glucose tolerance or known type 2
not fully correlate with the degree of visceral fat. CT, diabetes, hypertriglyceridemia, low HDL cholesterol
MRI and DXA scanning provide more accurate methods and hypertension. There are several definitions of the
of assessing visceral fat volume. Studies using these metabolic syndrome, of which two are in common use:
techniques have confirmed the inverse relationship of the International Diabetes Federation (IDF) (Zimmet
testosterone with visceral adiposity (Seidell et al. 1990; et al. 2005) and the National Cholesterol Education
Couillard et al. 2000). Total testosterone levels correlate Programme-Adult Treatment Panel III (NCEP ATP III)

236
Chapter 11: Obesity, diabetes and the metabolic syndrome

(Grundy et al. 2004) (Fig. 11.1). The main difference reduced risk of developing the metabolic syndrome
between the definitions is that, in the IDF, central (Blouin et al. 2005). These studies are important in that
obesity is an essential component, whereas it is not in they exclude obesity as a cause for the low testosterone
the NCEP ATP III. A joint statement has been released state; thus demonstrating that testosterone deficiency
in which these two definitions have been amalgamated may be a precursor to the development of obesity.
(see caption, Fig. 11.1; Alberti et al. 2009). Total testosterone has been reported to be 19%, free
The central biochemical defect which is common testosterone 11% and SHBG 18% lower in patients
between metabolic syndrome and type 2 diabetes is with metabolic syndrome compared to those without
insulin resistance. It is recognized that men with meta- it (Laaksonen et al. 2005). The association between low
bolic syndrome and impaired glucose tolerance are at testosterone and metabolic syndrome has also been
high risk of developing type 2 diabetes. Insulin resist- reported in other studies (Muller et al. 2005; Maggio
ance is the intermediary factor which promotes the et al. 2006). Furthermore, the greater the number of
development of hyperglycemia, hypertriglyceridemia, components of the metabolic syndrome, the lower the
low HDL cholesterol and hypertension. Factors which testosterone level and the greater the risk of clinical
contribute to the insulin resistance are central obesity, hypogonadism (Corona et al. 2006).
lack of exercise, genetic abnormalities and, more Men with metabolic syndrome have a higher preva-
recently identified, testosterone deficiency in men. lence of ED (26.7%) when compared to healthy controls
Insulin resistance is also associated with endothelial (13%) (Isidori et al. 2005a). The coexistence of testoster-
dysfunction (microalbuminuria), inflammation, small one deficiency with ED in men with metabolic syndrome
dense lipoproteins and a pro-thrombotic milieu. leads to more severe forms of ED (Corona et al. 2006).

11.3.2 Testosterone and the metabolic 11.3.3 Testosterone and type 2 diabetes
syndrome Numerous studies have consistently found that men with
Several epidemiological studies have shown that either type 2 diabetes have a high prevalence of low circulating
low total and/or free testosterone and SHBG are all testosterone levels. A meta-analysis of 21 studies up until
independent predictors for the subsequent clinical 2005, which included data from 3825 subjects, found that
development of the metabolic syndrome and for type total testosterone levels were, on average, 2.66 nmol/l
2 diabetes (Haffner et al. 1996; Stellato et al. 2000; Oh lower in men with diabetes compared to healthy controls
et al. 2002; Svartberg et al. 2004a; Laaksonen et al. (Ding et al. 2009). Until more recently it was assumed
2004; Selvin et al. 2007; Haring et al. 2009). These that this was mainly related to lower levels of SHBG that
include the Massachusetts Male Aging Study (MMAS), occur in men with states of insulin resistance, which
the Multiple Risk Factor Intervention Trial (MRFIT), include obesity, metabolic syndrome and type 2 diabetes.
Third National Health and Nutrition Examination Ding and colleagues in their meta-analysis found no
Survey (NHANES III), the Rancho Bernardo, Tromso significant decrease in SHBG, only a non-significant
and SHIP studies. The Rancho Bernardo Study, during reduction of 5.07 nmol/l (p = 0.15) in diabetic men.
an eight-year follow-up period, found an inverse cor- A landmark study, where the authors measured free
relation between baseline testosterone and fasting glu- testosterone by equilibrium dialysis in 103 men with
cose and insulin levels as well as glucose tolerance. diabetes (mean age 55 years), reported that one-third
Importantly, two studies, the MMAS (15-year had levels below the normal range (Dhindsa et al. 2004).
follow-up) and NHANES III, have found that the Another study, which assayed bioavailable testosterone
increased risk for metabolic syndrome and diabetes is by ammonium sulfate precipitation, found that 14%
also relevant for initially non-obese men (Kupelian et al. had levels below the normal range, with a further 36%
2006; Selvin et al. 2007). The NHANES III study in the lower 16% of the normal range (Kapoor et al.
reported a four-fold increased risk for the prevalence 2007a). The Third National Health and Nutrition
of diabetes in the lowest tertile of testosterone at base- Survey (NHANES III) reported that men in the lower
line, when the data were corrected for adiposity, age and tertile of free or bioavailable testosterone were approxi-
race/ethnicity. Conversely, when controlled for obesity, mately four times more likely to have diabetes than
the Quebec family study showed that men with higher those in the upper tertile with data adjusted for age,
total and bioavailable testosterone and SHBG had a obesity and ethnicity (Selvin et al. 2007).

237
Chapter 11: Obesity, diabetes and the metabolic syndrome

Guidelines for the clinical diagnosis of hypogo- doses to maintain glycemic control (Haider et al.
nadism state that the patient should have symptoms 2007). This implies that there should be extra vigi-
as well as biochemical evidence of testosterone defi- lance, for those men with diabetes, in the early phase
ciency. The first study to evaluate the prevalence of after commencing ADT (Levine et al. 2010). Andro-
hypogonadism in a population of men with type 2 gen deprivation therapy for prostate cancer produces
diabetes was published in 2007 (Kapoor et al. 2007a). a severe state of hypogonadism which increases cen-
This was a cross-sectional study of 355 men with type tral adiposity and percentage body fat, and decreases
2 diabetes who were >30 years of age, recruited lean mass (Smith et al. 2001; 2002; 2006). The change
mainly from the district retinopathy screening service in body fat is mainly subcutaneous and not visceral
as well as the hospital diabetic clinic. The prevalence fat, which is atypical for the metabolic syndrome
of hypogonadal men with a total testosterone level (Faris and Smith 2010). These findings all suggest a
<8 nmol/l was 17%, with a further 25% with total specific role for testosterone in carbohydrate as well
testosterone between 8 and 12 nmol/l. The prevalence as fat metabolism.
of hypogonadal men with a free testosterone level
< 255 pmol/l was 42%; bioavailable testosterone
< 2.5 nmol/l (lower limit of normal) was 14%, and 11.4 Interactions between the
29% had levels between 2.5 and 4 nmol/l. hypothalamic-pituitary-testicular axis
The most common form of classical hypogonad-
ism is Klinefelter syndrome. It has been shown that and adipose tissue
75% of men with this condition are undiagnosed in It is known that, first, a low testosterone state pro-
the general population (Bojesen et al. 2003). Further- motes an increase in fat deposition and that, second,
more, men with Klinefelter syndrome had a higher obesity impairs testosterone secretion. These two
risk of developing metabolic syndrome and type 2 important effects can lead to a vicious cycle between
diabetes than the normal population (Bojesen et al. obesity and hypogonadism. Several mechanisms are
2006). In addition, men with XSBMA, also called involved in this cycle, which mainly include the effects
Kennedy disease, a genetic polymorphism with excess of aromatase and adipocytokines. First, the Hypogo-
CAG repeats in exon 1 of the AR leading to a condi- nadal-Obesity Cycle was proposed to explain the sup-
tion associated with a relatively insensitive AR, which pressive effect of obesity on testosterone (Cohen
is associated with the development of diabetes, pro- 1999). More recently this has been developed to
vide further evidence of a link between impaired include the effects of adipocytokines known as
androgen status and diabetes. the Hypogonadal-Obesity-Adipocytokine Hypothesis
(Jones 2007). (Fig. 11.2).
11.3.4 Androgen deprivation therapy Aromatase activity which converts testosterone to
estradiol is primarily found in adipocytes and in par-
for prostate carcinoma ticular central fat deposits. The greater the degree of
The link between low testosterone and the increased adiposity, the higher the aromatase activity. Aroma-
risk of developing diabetes is supported by the effects tase inhibitors have been shown to increase testoster-
of ADT for prostatic carcinoma. A large follow-up one production in obese men (Zumoff et al. 2003;
study of 73 196 men treated with ADT, when com- Loves et al. 2008). The fact that clomifene, an anti-
pared to men with prostate cancer under surveillance, estrogen which works at the hypothalamic level, also
reported an increased risk of incident diabetes, myo- increases testosterone confirms the importance of the
cardial infarction and cardiovascular disease (Keating inhibitory action of estrogens on the hypothalamic-
et al. 2006). pituitary-testicular axis (Guay et al. 1995).
Fasting insulin levels rise and insulin sensitivity Adipose tissue is the largest endocrine organ,
falls, with an elevation in HbA1c, after initiation of which produces several hormones including pro-
ADT (Smith et al. 2006; Haider et al. 2007). A small inflammatory cytokines. Metabolic syndrome and
study of men with type 2 diabetes on insulin therapy, type 2 diabetes are recognized as pro-inflammatory
treated with ADT for advanced prostate cancer, states. Increased circulating free fatty acids activate
showed marked deterioration in their diabetic control NF-kB (nuclear factor kappa-light-chain-enhancer of
and required significant increases in their insulin activated B cells) pathways which then promote

238
Chapter 11: Obesity, diabetes and the metabolic syndrome

(4)

Estradiol

Hypothalamic-
TNF-
pituitary axis
IL-6

+
Leptin
(3)

Insulin
LH pulse
resistance
+ amplitude

(1)
Adipocyte number Testis
and size Aromatase

Adipose
Triglyceride tissue Testosterone
uptake

Lipoprotein lipase
activity

(2)
Fig. 11.2 The Hypogonadal-Obesity-Adipocytokine Hypothesis. (1) High aromatase activity in adipocytes converts testosterone to estradiol.
Reduced tissue testosterone facilitates triglyceride storage in adipocytes by allowing (2) increased lipoprotein lipase activity and stimulating
pluripotent stem cells to mature into adipocytes. (3) Increased adipocyte mass is associated with greater insulin resistance. (4) Estradiol and
adipocytokines TNF-a, IL-6 and leptin (as a result of leptin resistance in human obesity) inhibit the hypothalamic-pituitary-testicular axis
response to decreasing androgen levels (blue arrows). Orange arrow, hypogonadal obesity cycle; green arrow, low testosterone promotes the
formation of adipocytes from pluripotent stem cells; +, positive effect; , negative effect. See plate section for color version. (From Jones 2010).

cytokine release. Pro-inflammatory cytokines have a testosterone, there is no reciprocal increase in LH


major pathological role in atherogenesis and plaque levels (Strain et al. 1988; Zumoff et al. 2003). Men
instability. The pro-inflammatory cytokines or adipo- with insulin resistance and obesity have an impaired
cytokines involved are tumor necrosis factor a (TNF- Leydig cell response to exogenous gonadotropins (Pit-
a), interleukin 1 (IL-1), interleukin 6 (IL-6), IL-1b, teloud et al. 2005a).
plasminogen activator inhibitor 1 (PAI-1) and serum Kisspeptin is a peptide involved in the negative
amyloid A. Tumor necrosis factor a, IL-1b and IL-6 feedback of gonadal steroids on GnRH release from
are all known to suppress the hypothalamic-pituitary- the hypothalamus (George et al. 2010). Kisspeptin is
testicular axis (Jones and Kennedy 1993). The hor- produced in the rat arcuate and anterior periventri-
mone/cytokine leptin is exclusively synthesized and cular nuclei, and stimulates GnRH neurons via acti-
secreted by adipocytes. In human obesity, leptin vation of the KISS1 receptor. Kisspeptin stimulates
inhibits the hypothalamic release of GnRH. Further- the release of LH, FSH and testosterone. There is
more, leptin inhibits the action of hCG on the Leydig evidence from animal studies that estrogens and also
cell (Isidori et al. 1999). This explains why the com- leptin resistance inhibit the neuronal release of kis-
monest form of hypogonadism, hypogonadotropic speptin. These combined actions impair the homeo-
hypogonadism with low or normal LH levels, is static response to upregulate testosterone production
observed in men with central obesity. However, and in turn compensate for the increased breakdown
although weight loss can be associated with a rise in of testosterone to estradiol by adipose tissue.

239
Chapter 11: Obesity, diabetes and the metabolic syndrome

A persistent state of testosterone deficiency HDL-C is independently correlated positively with


promotes an increase in the amount of fat. It has been testosterone and estradiol (Van Pottelbergh et al.
shown that a low testosterone milieu results in 2003; Stanworth et al. 2011). Animal work has shown
favoring pluripotent stem cells to mature into adipo- that testosterone replacement in the testicular femin-
cytes as opposed to myocytes (Singh et al. 2003). ized mouse, which has an inactive AR, leads to an
Evidence is also accruing that a low testosterone state increase in HDL-C (Nettleship et al. 2007). This raises
facilitates the synthesis and storage of triglycerides in the hypothesis that HDL-C levels are related to estra-
fat cells. Adipocytes are known to express ARs diol and may be independent of the AR.
(Bjorntorp 1996). Testosterone has been shown to Androgen receptor sensitivity may be more
have a direct effect on cultured adipocytes and adi- important clinically in the presence of co-morbidities.
pose tissue, which implies that it has effects on intra- As described, testosterone levels are lower in a dia-
cellular lipid metabolism and potentially also has betic population, so a fall in testosterone in an indi-
effects on lipolysis and lipogenesis (Blouin et al. vidual with an insensitive receptor could give rise to
2008). Testosterone may also increase lipolysis by symptoms of hypogonadism, occurring with testos-
increasing the number of b-adrenergic receptors (De terone levels in the lower or even mid-normal range.
Pergola 2000).
Lipoprotein lipase, which resides on the extracel- 11.6 Mechanisms of testosterone
lular surface of adipocytes as well as being present in
the circulation, breaks down circulating triglycerides action on insulin sensitivity/resistance
to fat. Fatty acids are then taken up into the adipocyte Insulin resistance is recognized as the central bio-
and then converted back into triglycerides for storage. chemical defect in men with metabolic syndrome
Testosterone inhibits lipoprotein lipase activity; there- and/or type 2 diabetes. The presence of insulin resist-
fore, when testosterone is reduced this promotes tri- ance in type 2 diabetes is also known to be a major
glyceride storage. The increased rate of fat deposition cardiovascular risk factor, equivalent in degree to
also stimulates the transformation of immature pre- smoking (Bonora et al. 2002). In clinical practice the
adipocytes into mature cells. major cause of insulin resistance is central obesity;
however, lack of exercise and genetic factors are also
11.5 Role of the androgen receptor involved. The volume of central fat is directly propor-
tional to the degree of insulin resistance; whereas
polymorphism there is no relationship with subcutaneous fat.
It has been demonstrated that the CAG repeat poly- Population studies of healthy men have demon-
morphism in exon 1 of the androgen receptor influ- strated an inverse relationship between insulin and
ences the actions of testosterone in healthy men and testosterone levels (Simon et al. 1992; Tibblin et al.
those with Klinefelter syndrome (Zitzmann 2009). 1996). Androgen deprivation therapy for the treat-
There is recent evidence that this polymorphism ment of prostate carcinoma results in an increase in
modulates the effect of testosterone in men with type fasting insulin levels within three months with GnRH
2 diabetes (Stanworth et al. 2008; 2011). Although therapy (Smith et al. 2001; Dockery et al. 2003) and
men with type 2 diabetes had a distribution of CAG orchidectomy (Xu et al. 2002). The Rancho Bernardo
repeats similar to that found in a healthy population, Study found a significant inverse relationship of low
testosterone and LH levels were found to be signifi- baseline testosterone with follow-up fasting and post-
cantly increased in men with a more insensitive AR. challenge glucose and insulin levels and HOMA-IR
Importantly, less sensitive ARs are associated with (Homeostatic Model Assessment of Insulin Resist-
higher waist circumference, BMI, serum leptin and ance) eight years later (Oh et al. 2002).
systolic blood pressure. Sex hormone-binding globulin levels are inde-
High-density lipoprotein cholesterol was found to pendently inversely correlated with insulin resistance
be positively associated with increasing AR sensitivity. (Birkeland et al. 1993). This led to the presumption
The difficulty in evaluating this as a close association that it was the mechanism by which testosterone
is subject to a potential paradox. It is known that an levels were low in insulin-resistant states. Several
atherogenic lipid profile is associated with low testos- studies have shown that the bioavailable and free-
terone and estradiol. In healthy and diabetic men, testosterone fractions, which are independent of

240
Chapter 11: Obesity, diabetes and the metabolic syndrome

SHBG, correlate with insulin sensitivity (Andersson conversion to testosterone by 5a-reductase in rats)
et al. 1994; Endre et al. 1996; Simon et al. 1997). It is increased Glut-4 protein expression and translocation
important therefore to recognize that testosterone to the membrane (Sato et al. 2008). This study also
and SHBG are independently associated with insulin found that testosterone increased phosphorylation of
sensitivity (Kapoor et al. 2005, for review). Statin Akt and protein kinase C both key steps in the
therapy decreases SHBG and total testosterone but regulation of Glut-4 signaling pathways. These effects
has no effect on free or bioavailable levels (Stanworth were blocked by a DHT inhibitor, suggesting that
et al. 2009). These findings underline the need to local conversion of testosterone to DHT was essential
assess free or bioavailable testosterone status clinic- for this effect to occur. In addition, they reported that
ally, especially in borderline cases. testosterone increased activity of phosphofructoki-
The mechanisms by which testosterone promotes nase and hexokinase, key enzymes involved in
insulin sensitivity are not fully elucidated. The three glycolysis. Testosterone has also been shown to stimu-
tissues which account for the majority of the bodys late PI-3 kinase in osteoblasts, which increases Akt
insulin sensitivity are muscle, liver and fat (mainly phosphorylation (Kang et al. 2004).
central fat depots). Low testosterone states are associ- Mitochondrial dysfunction is also associated with
ated with increased adiposity, which in turn leads to insulin resistance. A study of 60 men using the hyper-
decreased insulin sensitivity. Changes in the sex ster- insulinemic-euglycemic clamp compared insulin
oid environment induced in healthy men by the aro- resistance with testosterone status (Pitteloud et al.
matase inhibitor letrozole, with or without estradiol 2005b). Testosterone levels correlated positively with
patches, demonstrated that testosterone increased insulin sensitivity, aerobic capacity (VO2max) and
insulin sensitivity and postprandial glucagon-like expression of genes involved in mitochondrial oxida-
peptide 1 (GLP-1) and decreased the postprandial rise tive phosphorylation from skeletal muscle biopsies.
in triglycerides (Lapauw et al. 2010). Interleukin 6 and The largest difference in mitochondrial gene expres-
TNF-a are known to promote insulin resistance. sion was found with ubiquinol cytochrome c reduc-
Acute withdrawal of testosterone from men with tase binding protein (UQCRB), showing a 20%
hypogonadism led to a relatively rapid reduction in reduction in tissue from men with type 2 diabetes
insulin sensitivity over a two-week period (Yialamas and impaired glucose tolerance.
et al. 2007). This finding implies that other mechan-
isms apart from body fat are involved.
In human type 2 diabetes, evidence shows that 11.7 Effect of testosterone
insulin resistance is associated with a reduction in replacement on insulin sensitivity
insulin-stimulated muscle glycogen synthesis, which
occurs as a result of a defect in insulin-stimulated 11.7.1 Obesity
transport activity. Studies have found that this can Marin and colleagues were the first to discover that
be ascribed to increases in lipid metabolites, which testosterone administration lowered plasma insulin
include fatty acyl CoA and diacylglycerol, which lead and glucose disposal over three months (Marin et al.
to stimulation of a serine/threonine kinase cascade, 1992a). However, a supraphysiological dose of testos-
which in turn produces defects in insulin signaling terone resulted in a reduction in glucose tolerance.
through serine/threonine phosphorylation of the The beneficial effects on insulin sensitivity were more
insulin receptor substrate 1 (IRS-1) in muscle (Sato pronounced in the men with lower testosterone levels.
et al. 2008). In liver a similar mechanism occurs, A decrease in waist/hip ratio was observed, but no
where increased diacylglycerol activates protein effect on body mass. Muscle strength, the fractional
kinase C, which results in reduced insulin-stimulated velocity of glycogen synthase and the diameter of type
tyrosine phosphorylation of IRS-2. There are few IIB fibers increased. Another study followed 23
published papers which have investigated potential middle-aged abdominally obese men for eight months
mechanisms by which testosterone increases insulin (Marin et al. 1992b). Testosterone therapy improved
sensitivity. insulin resistance as demonstrated by using the eugly-
Glut-4 is the major glucose transporter in muscle cemic-hyperinsulinemic clamp. A fall in blood glu-
and fat but is not present in liver. Testosterone and cose, diastolic blood pressure and cholesterol was also
DHEA in muscle (the latter probably by local detected.

241
Chapter 11: Obesity, diabetes and the metabolic syndrome

11.7.2 Metabolic syndrome and/or type 2 over 30 weeks in a randomized, double-blind, pla-
cebo-controlled trial in 184 men with metabolic syn-
diabetes drome and hypogonadism (Kalinchenko et al. 2010).
Kapoor and co-workers published the first study of Testosterone undecanoate 1000 mg was administered
TRT in hypogonadal men with type 2 diabetes in 2006 at 6 and 18 weeks by slow im injection. Although this
(Kapoor et al. 2006). This was a randomized, double- study included approximately one-third of subjects
blind, placebo-controlled, crossover trial with subjects with type 2 diabetes, results given were cumulated
assigned to either 200 mg im mixed testosterone from the total study population. The study did find
esters (24 mg testosterone propionate, 48 mg testos- that HOMA-IR decreased (p = 0.04).
terone phenylpropionate, 48 mg testosterone iso-
caproate, 80 mg testosterone undecanoate
Sustanon) every fortnight, or placebo every two
11.8 Effect of testosterone on
weeks for three months, with a one-month washout components of the metabolic
phase before crossing over for the final three months. syndrome and type 2 diabetes
This study found that insulin resistance as assessed by
HOMA-IR improved significantly with testosterone 11.8.1 Central adiposity and body
substitution. composition
A larger multicenter European study (the TIMES2
Testosterone substitution in men with hypogonadism
study) has confirmed that testosterone improved
is well known to improve body composition, with a
insulin resistance as assessed by HOMA-IR by 15%
decrease in fat and an increase in lean mass (Wang
after 6 months and was maintained after 12 months in
et al. 2000). Testosterone replacement in metabolic
the total population (Jones et al. 2011) (Fig. 11.3).
syndrome and/or type 2 diabetes has been shown to
This study recruited 220 men with either metabolic
reduce central adiposity in all but one trial, BMI in one
syndrome and/or type 2 diabetes aged  40 years,
(which involved an exercise component) and body fat
with symptoms of hypogonadism and either a morn-
content in another study (Kapoor et al. 2006; Heu-
ing total testosterone <11 nmol/l and/or a free testos-
felder et al. 2009; Kalinchenko et al. 2010; Jones et al.
terone <255 pmol/l. Subjects received either 60 mg
2011). Testosterone decreases serum leptin, which
2% testosterone gel or placebo. The testosterone dose
reflects body fat content (Kapoor et al. 2007b).
was titrated to give a testosterone level >17 nmol/l
(dose range 2080 mg/day) with dummy dose adjust-
ments in the placebo group. Changes in diabetes, lipid 11.8.2 Hyperglycemia
lowering, anti-obesity and hypertensive drugs were Hemoglobin A1c is the principal and gold standard
prohibited in the first 6 months unless crucial for test to assess glycemic control in people with type 2
clinical care. Between 6 and 12 months drug alter- diabetes. The UKPDS (United Kingdom Prospective
ations were allowed for ethical reasons. Diabetes Study) in type 2 diabetes showed that a
In the group of men with type 2 diabetes (n = 130), reduction in HbA1c had major benefits in reducing
testosterone therapy significantly reduced insulin the risk of both the microvascular (retinopathy, neph-
resistance by 15%, whereas a similar reduction was ropathy, neuropathy) and macrovascular (myocardial
observed in those men with metabolic syndrome infarction, stroke, peripheral vascular disease and
(n = 162), who showed a similar fall in insulin resist- sudden death) complications of type 2 diabetes
ance but approached significance, p = 0.0690, after 6 (Straton et al. 2000). Any intervention in lifestyle,
months and with p = 0.054 after 12 months. It is pharmacological or other method, that lowers HbA1c
important to recognize the high proportion of men would therefore have an important effect on out-
with diabetes who were already treated with metfor- comes in relation to these complications.
min. Metformin is known to improve insulin sensi- Two clinical trials have shown that testosterone
tivity by approximately 15%, so that the additional replacement in diabetic men lowers HbA1c. The first
benefit of testosterone would be in addition to the was the study of Kapoor and co-workers which
effects of this drug. reported a fall of HbA1c of 0.37% over three months,
The Moscow study examined the effect of depot with a fall in fasting blood glucose of 1.6 nmol/l
im testosterone undecanoate on insulin resistance (Kapoor et al. 2006). No other study has shown

242
Chapter 11: Obesity, diabetes and the metabolic syndrome

(A) All patients (D) All patients


20 0.6
0.5
Mean (95% CI) percentage change

15
from baseline in HOMA-IR (%)

from baseline in HbA1c (%)


0.4
10

Mean (95% CI) change


0.3
5
0.2
0
0.1
5 p = 0.372
0
10 0.1 p = 0.483
15 0.2 p = 0.056
20 0.3
p = 0.006 p = 0.120
25 p = 0.018 0.4
Baseline 6 months 12 months Baseline 3 months 6 months 9 months 12 months
Phase 1 Phase 2 Phase 1 Phase 2 Phase 2
Placebo n = 106 n = 106 Placebo n = 79 n = 76 n = 64 n = 59
Testosterone n = 99 n = 99 Testosterone n = 79 n = 71 n = 62 n = 49

(B) Patients with type 2 diabetes (E) Patients with type 2 diabetes
25 0.8
20
Mean (95% CI) percentage change

0.6
from baseline in HOMA-IR (%)

from baseline in HbA1c (%)


15
Mean (95% CI) change 0.4
10
5 0.2
0
0
5 p = 0.916 p = 0.566
10 0.2

15 0.4 p = 0.035

20 p = 0.057
0.6
p = 0.010
25 p = 0.049
Baseline 3 months 6 months 9 months 12 months
Baseline 6 months 12 months Phase 1 Phase 2 Phase 2
Phase 1 Phase 2 Placebo n = 48 n = 44 n = 38 n = 35
Placebo
n = 66 n = 66 n = 51 n = 46 n = 42 n = 31
Testosterone Testosterone
n = 64 n = 64

(C) Patients with MetS (F) Patients with MetS


20 0.6
0.5
Mean (95% CI) percentage change

15
from baseline in HOMA-IR (%)

0.4
from baseline in HbA1c (%)

10
Mean (95% CI) change

0.3
5
0.2
0
0.1
5 p = 0.429
0
10
0.1 p = 0.721
15 p = 0.106
0.2
20
p = 0.054 0.3
25 p = 0.069 p = 0.125
Baseline 6 months 12 months 0.4 Baseline 3 months 6 months 9 months 12 months
Phase 1 Phase 2 Phase 1 Phase 2 Phase 2
Placebo n = 82 n = 82 Placebo n = 63 n = 63 n = 52 n = 49
Testosterone n = 80 n = 80 Testosterone n = 64 n = 62 n = 54 n = 43

Fig. 11.3 Effect of TRT on insulin resistance (assessed by HOMA-IR) and HbA1c in hypogonadal men with metabolic syndrome (MetS) and/or
type 2 diabetes. Mean (95% CI) percentage change from baseline in HOMA-IR (intention-to-treat population, last observation carried forward)
and change from baseline HbA1c (intention-to-treat population, study completers) among all patients (A and D) and patients with type 2
diabetes (B and F), and with metabolic syndrome (C and F). The p-values are reported for comparisons between treatment and placebo groups.
(From Jones et al. 2011).

243
Chapter 11: Obesity, diabetes and the metabolic syndrome

a significant fall in fasting blood glucose, which Longer-term studies have shown that, after an initial
may be due to the confounding issue of concomitant fall in HDL-C, levels return to baseline with time
therapies in diabetic management. A second study (Jones et al. 2011). The reasons for the differences
compared diet and exercise with diet, exercise between studies are unclear; however, it has been
and testosterone substitution, which was non- postulated that testosterone stimulation of reverse
placebo-controlled, in newly diagnosed drug-nave cholesterol transport may lead to increased consump-
men with type 2 diabetes. After 12 months, those tion of HDL-C.
men treated with testosterone had an HbA1c Although in many studies testosterone levels cor-
reduction of 1.3% compared to 0.5% for those treated relate inversely with triglycerides, trials of testoster-
with diet and exercise alone: a comparative benefit of one replacement have generally not shown a
0.8% with testosterone (Heufelder et al. 2009). significant reduction in blood levels. The Heufelder
The TIMES2 study reported a significant reduc- study described above, in newly diagnosed drug-nave
tion in HbA1c (0.45%) compared to placebo after hypogonadal men with diabetes treated over 12
nine months treatment. This, however, was observed months, reported lowering in triglycerides (Heufelder
in phase 2 of the study where, for ethical reasons, et al. 2009). No effect has been observed in other
primary care physicians were allowed, where clinically studies involving diabetes or metabolic syndrome.
indicated, to alter hypoglycemic drugs. The fact that
changes occurred in both treatment and placebo
groups and were statistically significant does suggest 11.8.4 Hypertension
a potential therapeutic effect, but this cannot be cat- Hypotestosteronemia is associated with hypertension,
egorically stated. It is important however to recognize controlled or not (Khaw and Barrett-Connor 1988),
that the primary inclusion criterion for this study was but this could be related to a higher incidence of
hypogonadism, and subjects were not selected for obesity in men with this condition (Svartberg et al.
poor diabetic control. 2004b). Furthermore, an inverse relationship exists
between testosterone and diastolic blood pressure
(Bocchi et al. 2008; Jones and Saad 2009). Some
11.8.3 Dyslipidemia studies have found that testosterone reduces diastolic
The majority of cross-sectional studies have found and, less frequently, systolic blood pressure (Marin
that low testosterone is associated with an atherogenic et al. 1993; Kalinchenko et al. 2010; Jones 2010).Tes-
lipid profile, with high total and LDL cholesterol and tosterone and anabolic steroid, respectively, is known
triglycerides and low HDL-C, although some studies to increase blood pressure in animal studies and
disagree (Isidori et al. 2005b). The relationship humans (Jones 2010). Hence blood pressure should
between testosterone and HDL-C is complex and be monitored in TRT.
has not yet been fully elucidated. High-density lipo-
protein cholesterol protects against atherosclerotic
plaque progression, and levels are generally lower in 11.8.5 Inflammation
men. The major function of HDL-C is the reverse Low testosterone is associated with increased circu-
transport of cholesterol from the periphery to the lating IL-6 and C-reactive protein (CRP) in type 2
liver for excretion. In healthy and diabetic men tes- diabetes (Kapoor et al. 2007b). Testosterone replace-
tosterone positively correlates with HDL-C and its ment failed to suppress IL-1b, IL-6 and TNF-a, but
major constituent, apolipoprotein A1 (Van Pottel- reduced adiponectin and leptin in this study. Testos-
bergh et al. 2003; Stanworth et al. 2011). There is terone has been shown to decrease TNF-a and
evidence from animal studies that supports a hypoth- increase IL-10 in men with coronary heart disease
esis that testosterone conversion to 17b-estradiol (Malkin et al. 2004). In the Moscow study, testoster-
stimulates ERa-mediated production of HDL-C (Net- one substitution reduced TNF-a, IL-1b and CRP,
tleship et al. 2007). whereas a fall in IL-6 approached significance, but
The majority of studies have either shown no treatment had no effect on IL-10, an anti-atherogenic
effect or a small decrease in HDL-C with testosterone cytokine (Kalinchenko et al. 2010). Testosterone
therapy. However, trials with testosterone replacement has been shown to inhibit TNF-a, IL-
undecanoate depot report an increase in HDL-C. 1b and IL-6 from cultured peripheral blood

244
Chapter 11: Obesity, diabetes and the metabolic syndrome

monocytes in vitro, from androgen-deficient men 11.11 Clinical implications


with type 2 diabetes (Corrales et al. 2006).
International clinical guidelines now recommend that
serum testosterone should be measured in men with
11.9 Cholesterol and lipoproteins type 2 diabetes mellitus with symptoms suggestive of
Although cholesterol is not included in the definition testosterone deficiency (Wang et al. 2008). The clin-
of metabolic syndrome, this substance plays an ical diagnosis does need careful and rigorous assess-
important role in atherogenesis in men with these ment before a decision to treat can be made. There is
conditions. Total and LDL cholesterol are both inde- now evidence that testosterone replacement can
pendently and negatively correlated with testosterone improve sexual health and the response to phospho-
(Isidori et al. 2005b). Meta-analyses of clinical trials diesterase-5 inhibitors. Evidence is accumulating to
have shown that testosterone replacement reduces support a beneficial action of testosterone on insulin
total and LDL cholesterol. Intervention studies have sensitivity and cardiovascular risk factors (Wang et al.
found that testosterone replacement significantly 2011). Larger randomized, placebo-controlled trials
lowers cholesterol and LDL cholesterol between 0.25 are still required to verify these benefits and to deter-
and 0.5 mmol/l even in men already treated with mine whether these effects persist over a longer
statins (Jones and Saad 2009). period of time.
Lipoprotein (a) (Lpa) is the strongest independent
risk factor for cardiovascular disease. The function of 11.12 Key messages
Lpa is unknown. Recently the European Atheroscler-  There is a high prevalence of hypogonadism in
osis Society has advised that Lpa, which is mainly men with metabolic syndrome and type 2
genetically determined, be measured in men who have diabetes.
moderate or high cardiovascular risk. Testosterone  Low testosterone and SHBG in healthy men
substitution decreases Lpa in hypogonadal men with are independent risk factors for the future
and without diabetes (Zmuda et al. 1996; Jones et al. onset of metabolic syndrome and type
2011). There is currently no evidence that reducing 2 diabetes.
Lpa has any effect on overall cardiovascular risk.  The interrelationships between adipose
tissue and the hypothalamic-pituitary axis
suppress the homeostatic mechanism, leading
11.10 Erectile dysfunction to impaired testosterone production and
Erectile dysfunction is common in men with meta- eventually to a state of hypogonadotropic
bolic syndrome (26%) and in type 2 diabetes (up to hypogonadism.
70%) (Wang et al. 2011). Erectile dysfunction is mul-  Insulin resistance is the central biochemical
tifactorial in diabetic men, with vasculopathy, auto- defect in metabolic syndrome and type
2 diabetes, which adversely affects cardiovascular
nomic neuropathy, hypogonadism and psychological
risk factors, resulting in the increased number of
factors being involved. Erectile dysfunction may be
cardiovascular events and mortality in these
the first clinical sign of cardiovascular disease and the conditions.
herald of future cardiovascular events including myo-  Testosterone has a key role in promoting
cardial infarction. Early diagnosis provides a window insulin sensitivity via actions on carbohydrate
of opportunity, to improve lifestyle issues and medi- and fat metabolism as well as insulin signaling
cation, to reduce cardiovascular risk and ultimately pathways.
events and death.  Short-term studies up to one year in duration
The severity of ED is greater the lower the testos- have shown that TRT improves insulin
terone level in type 2 diabetes (Kapoor et al. 2007c). resistance, but not all have had consistent
Low testosterone is associated with a failure to benefits to other components of the metabolic
syndrome.
respond to sildenafil, and replacement can convert
 Erectile dysfunction may be the first symptom
non-responders to responders. Testosterone substitu-
of vascular disease, and men should be treated
tion improves libido and the International Index of to reduce cardiovascular risk. There is a high
Erectile Function score in hypogonadal men with prevalence of testosterone deficiency in men
metabolic syndrome and/or type 2 diabetes with ED.
(Jones et al. 2011).

245
Chapter 11: Obesity, diabetes and the metabolic syndrome

 Testosterone replacement in hypogonadal men  The clinical diagnosis of hypogonadism requires


with metabolic syndrome and/or type 2 diabetes careful assessment of symptoms, and
improves sexual health. biochemical testosterone deficiency.

11.13 References registry study. J Clin Endocrinol


Metab 88:622628
De Pergola G (2000) The adipose tissue
metabolism: role of testosterone and
Alberti KGMM, Eckel RH, Grundy SM, dehydroepiandrosterone. Int J Obes
Zimmet PZ, Cleeman JI, Donato Bojesen A, Kristensen K, Birkeback NH
(2006) The metabolic syndrome Relat Metab Disord 24 (Suppl 2):
KA, Fruchart J-C, James WPT, Loria S59S63
CM, Smith SC (2009) Harmonizing is frequent in men with
the metabolic syndrome. Circulation Klinefelters Syndrome. Diabetes Dhindsa S, Prabhakar S, Sethi M,
120:16401645 Care 29:15911598 Bandyo Padhyay A, Chaudhuri A,
Dandona P (2004) Frequent
Allan CA, McLachlan RI (2010) Bonora E, Formentini G, Calcaterra F,
occurrence of hypogonadotrophic
Androgens and obesity. Curr Lombardi S, Marini F, Zenari L,
hypogonadism in type 2 diabetes.
Opin Endocrinol Diabetes Obes Saggiani F, Poli M, Perbellini S,
J Clin Endocrinol Metab 89:54625468
17:224232 Raffaelli A, Cacciatori V, Santi L,
Targher G, Bonadonna R, Muggeo Ding EL, Song Y, Manson JE, Hunter
Andersson B, Marin P, Lissner L, DJ, Lee CC, Rifai N, Buring JE,
M (2002) HOMA-estimated insulin
Vermeulen A, Bjorntorp P (1994) Gaziano JM, Liu S (2009) Sex
resistance is an independent
Testosterone concentrations in hormone-binding globulin and
predictor of cardiovascular disease
women and men with NIDDM. risk of type 2 diabetes in women
in type 2 diabetic subjects:
Diabetes Care 17:405411 and men. N Engl J Med 361:
prospective data from the Verona
Birkeland KI, Hanssen KF, Torjesen Diabetes Complications Study. 11521163
PA, Vaaler S (1993) Level of sex Diabetes Care 25:11351141 Dockery F, Bulpitt CJ, Agarwal S,
hormone binding globulin is Donaldson M, Rajkumar C (2003)
Cohen P (1999) The hypogonadal-
positively correlated with insulin Testosterone suppression in men
obesity-cycle. Med Hypothesis
sensitivity in men with type 2
52:4951 with prostate cancer leads to an
diabetes. J Clin End Metab increase in arterial stiffness and
76:275278 Corona G, Mannucci E, Schulman C, hyperinsulinaemia. Clin Sci
Bjorntorp P (1996) The regulation of Petrone L, Mansani R, Cilotti A, 104:195201
adipose tissue distribution in Balercia G, Chiarini V, Forti G,
Maggi M (2006) Psychobiologic Endre T, Mattiasson I, Berglund G,
humans. Int J Obes Relat Metab Hulthen UL (1996) Low
Disord 20:291302 correlates of the metabolic
syndrome and associated sexual testosterone and insulin resistance
Blouin, K, Despres JP, Couillard C, dysfunction. Eur Urol 50:595604 in hypertension prone men. J Hum
Tremblay A, Prudhomme D, Hypertens 10:755761
Bouchard C, Tchernuf A (2005) Corrales JJ, Almeida M, Burgo R,
Mories MT, Miralles JM, Orfao A Faris JE, Smith MR (2010) Metabolic
Contribution of age and declining sequelae associated with androgen
androgen levels to features of the (2006) Androgen replacement
therapy depresses the ex-vivo deprivation therapy for prostate
metabolic syndrome in men. cancer. Curr Opin Endocrinol
Metabolism 54:10341040 production on inflammatory
cytokines by circulating antigen- Diabetes Obes 17:240246
Blouin K, Boivin A, Tchernof A (2008) presenting cells in aging type 2 George JT, Miller RP, Anderson RA
Androgens and body fat diabetic men with partial (2010) Hypothesis: kisspeptin
distribution. J Steroid Biochem Mol androgen deficiency. J Endocrinol mediates male hypogonadism in
Biol 108:272280 189:595604 obesity and type 2 diabetes.
Bocchi EA, Carvalho VO, Guimaraes Couillard C, Gagnon J, Bergeron J, Neuroendocrinology 91:
GV (2008) Inverse correlation Leon AS, Rao DC, Skinner JS, 302307
between testosterone and ventricle Wilmore JH, Desprs JP, Bouchard Grundy SM, Brewer HB Jr, Cleeman JI,
ejection fraction, hemodynamics C (2000) Contribution of body Smith SC Jr, Lenfant C (2004)
and exercise capacity in heart failure fatness and adipose tissue Definition of metabolic syndrome:
patients with erectile dysfunction. distribution to the age variation in report of the National Heart, Lung,
Int Braz J Urol 34:302310 plasma steroid hormone and Blood Institute/American Heart
Bojesen A, Juul S, Gravholt CH (2003) concemtrations in men: the Association conference on scientific
Prenatal and postnatal prevalence of HERITAGE Family Study. J Clin issues related to definition.
Klinefelters syndrome: a national Endocrinol Metab 85:10261031 Circulation 109:433438

246
Chapter 11: Obesity, diabetes and the metabolic syndrome

Guay AM, Bansal S, Heatley GJ (1995) analysis. Clin Endocrinol (Oxf) Kapoor D, Malkin CJ, Channer KS,
Effect of raising endogenous 63:381394 Jones TH (2005) Androgens,
testosterone levels in impotent men Isidori AM, Gianetta E, Greco EA, insulin resistance and vascular
with secondary hypogonadism: Gianfrilli D, Bonifacio V, Isidori A, disease in men. Clin Endocrinol
double blind placebo-controlled Lenzi A, Fabbri A (2005b) Effects of (Oxf) 63:239250
trial with clomiphene citrate. J Clin testosterone on body composition, Kapoor D, Goodwin E, Channer KS,
Endocrinol Metab 80:35463552 bone metabolism and serum lipid Jones TH (2006) Testosterone
Haffner SM, Valdez RA, Stern MP, profile in middle-aged men: a meta- replacement therapy improves
Katz MS (1993) Obesity, body fat analysis. Clin Endocrinol (Oxf) insulin resistance, glycaemic
distribution and sex hormones in 63:280293 control, visceral adiposity and
men. Int J Obes Relat Metab Disord Jones TH (2007) Testosterone hypercholesterolaemia in
17:643649 associations with erectile hypogonadal men with type 2
Haffner SM, Shaten J, Stern MP, Smith dysfunction, diabetes and the diabetes. Eur J Endocrinol
GD, Kuller L (1996) Low levels of metabolic syndrome. Eur Urol Suppl 154:899906
sex hormone binding globulin and 6:847857 Kapoor D, Aldred H, Clark S, Channer
testosterone predict the KS, Jones TH (2007a) Clinical and
Jones TH (2010) Testosterone
development of non insulin biochemical assessment of
deficiency: a risk factor for
dependent diabetes mellitus in men. hypogonadism in men with type 2
cardiovascular disease? Trends
Am J Epidemiol 143:889897 Endocrinol Metab 21:496503
diabetes: correlations with
bioavailable testosterone and
Haider A, Yassin A, Saad F, Shabsigh R Jones TH, Kennedy RL (1993) visceral adiposity. Diabetes Care
(2007) Effect of androgen deprivation Cytokines and hypothalamic- 30:911917
on glycaemic control and on pituitary function. Cytokine
cardiovascular risk factors in men Kapoor D, Clarke S, Channer KS, Jones
5:531538
with advanced prostate cancer with TH (2007b) The effect of
diabetes. Aging Male 10:189196 Jones TH, Saad F (2009) The effects of testosterone replacement therapy on
testosterone on risk factors for, and adipocytokines and C-reactive
Haring R, Volzke H, Felix SB, Schipf S, the mediators of, the atherosclerotic protein in hypogonadal men with
Dorr M, Rosskopf D, Nauck M, process. Atherosclerosis 207:318327 type 2 diabetes. Eur J Endocrinol
Schofl C, Wallaschofski H (2009)
Jones TH, Arver S, Behre HM, Buvat J, 156:595602
Prediction of metabolic syndrome
by low serum testosterone levels in Meuleman E, Moncada I, Morales Kapoor D, Clarke S, Channer KS, Jones
men. Results from the study of AM, Volterrani M, Yellowlees A, TH (2007c) Erectile dysfunction is
health in Pomerania. Diabetes Care Howell JD, Channer KS (2011) associated with low bioactive
58:20272031 Testosterone replacement in testosterone levels and visceral
hypogonadal men with type 2 adiposity in men with type 2
Heufelder AE, Saad F, Bunck MC, diabetes and/or metabolic diabetes. Int J Androl 30:500507
Gooren L (2009) Fifty-two-week syndrome (The TIMES2 Study).
treatment with diet and exercise Keating NL, OMalley AJ, Freedland SJ,
Diabetes Care 34:828837
plus transdermal testosterone Smith MR (2006) Diabetes and
reverses the metabolic syndrome Kalinchenko SY, Tishova YA, cardiovascular disease during
and improves glycemic control in Mskhalaya GJ, Gooren LJ, Giltay EJ, androgen deprivation therapy:
men with newly diagnosed type 2 Saad F (2010) Effects of testosterone observational study of veterans with
diabetes and subnormal plasma supplementation on markers of the prostate cancer. J Natl Cancer Inst
testosterone. J Androl 30:726733 metabolic syndrome and 102:3946
inflammation in hypogonadal men Khaw KT, Barrett-Connor E (1988)
Isidori AM, Caprio M, Stroll F, Moretti with the metabolic syndrome: the
C, Frajese G, Isidori A, Fabbri A Blood pressure and endogenous
double-blinded placebo-controlled testosterone in men: an inverse
(1999) Leptin and androgens in Moscow study. Clin Endocrinol
male obesity: evidence for leptin relationship. J Hypertens 6:329332
(Oxf) 73:602612
contribution to reduced androgen Khaw KT, Barrett-Connor E (1992)
levels J Clin Endocrinol Metab Kang HY, Cho CL, Huang KL, Wang Lower endogenous androgens
84:36733680 JC, Hu YC, Lin HK, Chang C, predict central adiposity in men.
Huang KE (2004) Nongenomic Am Epidemiol 2:675682
Isidori AM, Giannetta E, Gianfrilli D, androgen activation of
Greco EA, Bonifacio V, Aversa A, phosphatidylinositol3-kinase/Akt Kupelian V, Page ST, Araujo AB,
Isidori A, Fabbri A, Lenzi A (2005a) signalling pathway in MC3T3-E1 Travison TG, Bremner WJ,
Effects of testosterone on sexual osteoblasts. J Bone Miner Res McKinlay JB (2006) Low sex
function in men: results of a meta- 19:11811190 hormone-binding globulin, total

247
Chapter 11: Obesity, diabetes and the metabolic syndrome

testosterone, and symptomatic analysis. Hum Reprod Update of type 2 diabetes in older men and
androgen deficiency are associated 16:293311 women. Diabetes Care 25:5560
with development of the metabolic Maggio M, Lauretani F, Ceda GP, Pasquali R, Casimirri F, Cantobelli S,
syndrome in non-obese men. J Clin Bandinelli S, Basaria S, Ble A, Egan J, Melchionda N, Morselli Labate AM,
Endocrinol Metab 91:843850 Paolisso G, Najjar S, Jeffrey Fabbri R, Capelli M, Bortoluzzi L
Laaksonen DE, Niskanen L, Punnonen Metter E, Valenti G, Guralnik JM, (1991) Effect of obesity and body fat
K, Nyyssonen K, Tuomainen TP, Ferrucci L (2006) Association distribution on sex hormones and
Valkonen VP, Salonen R, Salonen between hormones and metabolic insulin in men. Metabolism
JT (2004) Testosterone and sex syndrome in older Italian men. 40:101104
hormone-binding globulin J Am Geriatr Soc 54:18321838 Pitteloud N, Hardin M, Dwyer AA,
predict the metabolic syndrome Malkin CJ, Pugh PJ, Jones RD, Kapoor Valassi E, Yialamas M, Elahi D,
and diabetes in middle-aged D, Channer KS, Jones TH (2004) Hayes FJ (2005a) Increasing insulin
men. Diabetes Care 27: The effect of testosterone resistance is associated with a
10361041 replacement on endogenous decrease in Leydig cell testosterone
Laaksonen DE, Niskanen L, Punnonen inflammatory cytokines and lipid secretion in men. J Clin Endocrinol
K, Nyyssonen K, Tuomainen TP, profiles in hypogonadal men. J Clin Metab 90:26362641
Valkonen VP, Salonen JT (2005) Endocrinol Metab 89:33133318 Pitteloud N, Mootha VK, Dwyer AA,
The metabolic syndrome and Marin P, Krotkiewski M, Bjorntorp P Hardin M, Lee H, Eriksson KF,
smoking in relation to (1992a) Androgen treatment of Tripathy D, Yialamas M, Groop L,
hypogonadism in middle-aged men: middle-aged, obese men: effects on Elahi D, Hayes FJ (2005b)
a prospective cohort study. J Clin metabolism, muscle and adipose Relationship between testosterone
Endocrinol Metab 90:712719 tissues. Eur J Med 1:329336 levels, insulin sensitivity, and
Lapauw B, Ouwens M, Hart LM, Marin P, Holmang S, Jonsson L, mitochondrial function in men.
Wuyts B, Holst JJ, TSjoen G, Sjostrom L, Kvist H, Holm G, Diabetes Care 28:16361642
Kaufman JM, Ruige JB (2010) Sex Lindstedt G, Bjorntorp P (1992b) Sato K, Iemitsu M, Aizawa K, Ajisaka R
steroids affect triglyceride handling, The effects of testosterone treatment (2008) Testosterone and DHEA
glucose-dependent insulinotropic on body composition and activate the glucose metabolism-
polypeptide, and insulin sensitivity: metabolism in middle-aged obese related signalling pathway in
a 1-week randomized clinical trial in men. Int J Obes Relat Metab Disord skeletal muscle. Am J Physiol
healthy young men. Diabetes Care 16:991997 Endocrinol Metab 294:E961E968
33:18311833 Marin P, Holmang S, Gustafsson C, Seidell JC, Bjorntorp P, Sjostrom L,
Levine GN, DAmico AV, Berger P, Jonsson L, Kvist H, Elander A, Eldh Kvist H, Sannerstedt R (1990)
Clark PE, Eckel RH, Keating NL, J, Sjostrom L, Holm G, Bjorntorp P Visceral fat accumulation in men is
Milani RV, Sagalowsky AI, Smith (1993) Androgen treatment of positively associated with insulin,
MR, Zakai N (2010) Androgen- abdominally obese men. Obes Res glucose, and C-peptide levels, but
deprivation therapy in prostate 1:245251 negatively with testosterone levels.
cancer and cardiovascular risk: a Muller M, Grobbee DE, den Tonkelaar Metabolism 39:897901
science advisory from the American I, Lamberts SW, van der Schouw YT Selvin E, Feinleib M, Zhang L,
Heart Association, American (2005) Endogenous sex hormones Rohrmann S, Rifai N, Nelson WG,
Cancer Society and American and metabolic syndrome in aging Dobs A, Basaria S, Golden SH, Platz
Urological Association: endorsed by men. Clin Endocrinol Metab. EA (2007) Androgens and diabetes
the American Society for Radiation 90:26182623 in men. Results from the Third
Oncology. Circulation 121:833840 National Health and Nutrition
Nettleship JE, Jones TH, Channer KS,
Loves S, Ruinemans-Koerts J, de Boer Jones RD (2007) Physiological Examination Survey (NHANES III).
H (2008) Letrozole once a week testosterone replacement therapy Diabetes Care 30:234238
normalizes serum testosterone in attenuates fatty streak formation Simon D, Preziosi P, Barrett-Connor E,
obesity-related male hypogonadism. and improves high-density Roger M, Saintpaul M, Nahoul K,
Eur J Endocrinol 158:741747 lipoprotein cholesterol in the Tfm Papoz L (1992) Interrelation
MacDonald AA, Herbison GP, Showell mouse: an effect independent of the between plasma testosterone and
M, Farquhar CM (2010) The impact classic androgen receptor. plasma-insulin in healthy adult
of body mass index on semen Circulation 116:24272434 men the Telecom study.
parameters and reproductive Oh JY, Barrett-Connor E, Wedick NM, Diabetologia 35:173177
hormones in human males: a Wingard DL (2002) Endogenous Simon D, Charles MA, Nahoul K,
systematic review with meta- sex hormones and the development Orssaud G, Kremski J, Hully V,

248
Chapter 11: Obesity, diabetes and the metabolic syndrome

Joubert E, Papoz L, Eschwege E associated with testosterone, of testosterone and estradiol in the
(1997) Association between plasma oestradiol and androgen receptor determination of cholesterol and
total testosterone and CAG repeat polymorphism in men lipoprotein profile in healthy
cardiovascular risk factors in with type 2 diabetes. Clin Endocrinol middle-aged men. Atherosclerosis
healthy adult men: the Telecom (Oxf) 74:624630 166:95102
Study. J Clin Endocrinol Metab Stellato RK, Feldman HA, Hamdy O, Wang C, Swerdloff RS, Iranmanesh A,
82:682685 Horton ES, McKinlay JB (2000) Dobs A, Snyder PJ, Cunningham G,
Singh R, Artaza JN, Taylor WE, Testosterone, sex hormone binding Matsumoto AM, Weber T, Berman
Gonzalez-Cadavid NF, Bhasin S globulin and the development of N; Testosterone Gel Study Group
(2003) Androgens stimulate type 2 diabetes in middle aged men. (2000) Transdermal testosterone gel
myogenic differentiation and inhibit Diabetes Care 23:490494 improves sexual function, mood,
adipogenesis in C3H 10T1/2 Strain GW, Zumoff B, Miller L, Rosner muscle strength and body
pluripotent cells through an W, Levit C, Kalin M, Hershcopf RJ, composition parameters in
androgen receptor-mediated Rosenfeld RS (1988) Effect of hypogonadal men. J Clin Endocrinol
pathway. Endocrinology 144: massive weight loss on Metab 85:28392853
50815088 hypothalamic-pituitary-gonadal Wang C, Nieschlag E, Swerdloff R,
Smith JC, Bennett S, Evans LM, function in obese men. J Clin Behre HM, Hellstrom WJ, Gooren
Kynaston HG, Parmar M, Mason Endocrinol Metab 66:10191023 LJ, Kaufman JM, Legros JJ,
MD, Cockcroft JR, Scanlon MF, Straton IM, Adler AI, Neil AW, Lunenfeld B, Morales A, Morley JE,
Davies JS (2001) The effects of Matthews DR, Manley SE, Cull CA, Schulman C, Thompson IM,
induced hypogonadism on arterial Hadden D, Turner RC, Holman RR Weidner W, Wu FCW (2008)
stiffness, body composition and (2000) Association of glycaemia Investigation, treatment and
metabolic parameters in males with with macrovascular and monitoring of late-onset
prostate cancer. J Clin Endocrinol microvascular complications of hypogonadism in males: ISA,
Metab 86:42614267 type 2 diabetes (UKPDS 35): ISSAM, EAU, EAA and ASA
Smith MR, Finkelstein JS, McGovern prospective observational study. recommendations. Eur J Endocrinol
FJ, Zietman AL, Fallon MA, BMJ 321:405412 159:507514
Shoenfeld DA, Kantoff PW (2002) Svartberg J, von Muhlen D, Sundsfjord Wang C, Jackson G, Jones TH,
Changes in body composition J, Jorde R (2004a) Waist Matsumoto AM, Nehra A,
during androgen deprivation circumference and testosterone Perelman MA, Swerdloff RS, Traish
therapy for prostate cancer. J Clin levels in community dwelling men. A, Zitzmann M, Cunningham G
Endocrinol Metab 87: The Tromso study. Eur J Epidemiol (2011) Low testosterone associated
599603 19:657663 with obesity and the metabolic
Smith MR, Lee H, Nathan DM (2006) Svartberg J, von Muhlen D, Schirmer H, syndrome contributes to sexual
Insulin sensitivity during combined Barrett-Connor E, Sundsfjord J, dysfunction and cardiovascular
androgen blockade for prostate Jorde R (2004b) Association of disease risk in men with
cancer. J Clin Endocrinol Metab endogenous testosterone with blood type 2 diabetes. Diabetes Care
91:13051308 pressure and left ventricular mass in 34:16691675
Stanworth RD, Kapoor D, Channer KS, men: the Tromso study. Eur Xu T, Wang X, Hou S, Zhu J, Zhang X,
Jones TH (2008) Androgen receptor J Endocrinol 159:6571 Huang X (2002) Effect of surgical
CAG repeat polymorphism is Tibblin G, Adlerberth A, Lindstedt G, castration on risk factors for
associated with serum testosterone Bjorntorp P (1996) The pituitary arteriosclerosis of patients with
levels, obesity and serum leptin gonadal axis and health in elderly prostate cancer. Chin Med J
in men with type 2 diabetes. men: a study of men born in 1913. 115:13361340
Eur J Endocrinol 159: Diabetes 45:16051609 Yialamas MA, Dwyer AA, Hanley E,
736746 Lee H, Pitteloud N, Hayes FJ (2007)
Tsai EC, Boyko EJ, Leonetti DL,
Stanworth RD, Kapoor D, Channer KS, Fujimoto WY (2000) Low serum Acute sex steroid withdrawal
Jones TH (2009) Statin therapy is testosterone level as a predictor of reduces insulin sensitivity in healthy
associated with lower total but not increased visceral fat in Japanese- men with idiopathic
bioavailable or free testosterone in American men. Int J Obes Relat hypogonadotrophic hypogonadism.
men with type 2 diabetes. Diabetes Metab Disord 24:485491 J Clin Endocrinol Metab 92:
Care 32:541546 Van Pottelbergh I, Braeckman L, De
42544259
Stanworth RD, Kapoor D, Channer KS, Bacquer D, De Backer G, Kaufman Zimmet P, Magliano D,
Jones TH (2011) Dyslipidaemia is JM (2003) Differential contribution Matsuzawa Y, Alberti G, Shaw J

249
Chapter 11: Obesity, diabetes and the metabolic syndrome

(2005) The metabolic polymorphism in andrology. Front Zumoff B, Miller LK, Strain GW
syndrome: a global public Horm Res 37:5261 (2003) Reversal of the
health problem and a new Zmuda JM, Thompson PD, Dickenson hypogonadotropic hypogonadism
definition. J Atheroscler Thromb R, Baussermann LL (1996) of obese men by the administration
12:295300 Testosterone decreases lipoprotein of the aromatase inhibitor
Zitzmann M (2009) The role of CAG (a) in men. Am J Cardiol testolactone. Metabolism
repeat androgen receptor 77:12441247 52:11261128

250
Chapter
Testosterone and erection

12 Mario Maggi and Hermann M. Behre

12.1 Introduction 251 12.7 Prevalence of testosterone


12.2 Physiology of erection 252 deficiency in patients with erectile
12.3 Direct effects of testosterone on penile dysfunction 259
growth and erection 252 12.8 Effects of sexual activity and
12.4 Association between androgen level and treatment of erectile dysfunction on
erectile activity in men with erectile testosterone 260
dysfunction 255 12.9 Conclusion 261
12.5 Effects of testosterone therapy on 12.10 Key messages 262
erection 257 12.11 References 262
12.6 Combined therapy with testosterone and
phosphodiesterase type 5 inhibitors in
patients with erectile dysfunction 257

12.1 Introduction and lifestyle factors are simultaneously present and


Erectile dysfunction (ED) is a multifactorial disorder, mutually interacting (Petrone et al. 2003). In other
and several emotional, physical and medical factors con- words, ED is a frustrating symptom deriving from a
tribute to the degree of dysfunction that significantly continuous spectrum of clinical pictures including phys-
impairs the patient and partners quality of life, having ical illness (organic component of ED), reaction to life
a detrimental effect on sexual and reproductive activity. stresses (intrapsychic component of ED) or an unhappy
A recent analysis of all population-based studies con- couple relationship (relational component of ED) (Pet-
ducted in the USA indicates that ED is indeed the most rone et al. 2003). We strongly believe that the male
common endocrine disorder in men (Golden et al. 2009). hormone, testosterone, plays a relevant role in determin-
A European survey shows that ED affects almost 30% of ing perturbations in all three aforementioned dimen-
men in an age-dependent manner (Corona et al. 2010a). sions of ED (organic, intrapsychic and relational).
In 1993, the NIH Consensus Development Panel on The prevalence and the severity of ED increase
Impotence defined ED as the inability to attain and/or with advancing age (Rosen et al. 2003; Corona et al.
maintain penile erection sufficient for satisfactory sexual 2004; 2010a; Lindau et al. 2007). In the Massachusetts
performance (NIH Consensus Conference 1993). Male Aging Study (Feldman et al. 1994), the propor-
Penile erection is a neurovascular event that can be tion of subjects with severe ED increased from 5% at
seen as an integrated feed-forward interaction within a the age of 40 years to 15% at the age of 70. Elderly
biological dimension (with its cardiovascular, neuronal patients are often affected by multiple organic
and hormonal determinants), an intrapsychic dimension diseases, which can interfere with sexual function-
(the individuals sexual identity and sense of well-being), ing. In particular, cardiovascular and metabolic dis-
and a marital dimension (the context for a sexual rela- eases with increased prevalence in the elderly
tionship). In each ED patient, biological, psychological (including CAD, myocardial infarction, hypertension,

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

251
Chapter 12: Testosterone and erection

hyperlipidemia, diabetes mellitus and peripheral vas- nervous system control the switch between muscle
cular disease) could play a crucial role in the patho- contraction and relaxation (Berridge 2008). Sympa-
genesis of many cases of age-related sexual thetic nerve activity is mainly responsible for main-
dysfunction (Feldman et al. 1994; Rosen et al. 2003; taining SMC in a contractile state (see Fig. 12.1A).
Corona et al. 2004; 2010a; Lindau et al. 2007). Also The release of noradrenalin (NA) from nerve endings
testosterone declines gradually as a function of aging; allows its binding to cognate receptors, which activate
however, its relationship with ED is still debated. Data two separate pathways within the cells, favoring
from the EMAS (European Male Aging Study), a increase in intracellular calcium and sensitization to
population-based study involving more than 3000 it (see Fig. 12.1A). Smooth muscle cell relaxation is
subjects from 8 European centers, showed an primarily driven by the activity of parasympathetic
unadjusted annual age decline for total testosterone and non-adrenergic, non-cholinergic (NANC) nerve
of 0.04 nmol/l per year (Wu et al. 2008). In line with endings, which control nitric oxide (NO) formation
the EMAS results, a significant age-dependent decline through NO synthase (NOS), present in both endothe-
also of total testosterone of 0.09 nmol/lyear was lial cells (eNOS) and neurons (nNOS) (see Fig. 12.1B).
observed in subjects with ED (Corona et al. 2011a). Nitric oxide diffuses into SMC and increases the
formation of cyclic GMP, an action actively counter-
acted by a series of phosphodiesterases (PDEs), the
12.2 Physiology of erection most important of which is PDE5. In human corpora
Erection can be regarded as a complex neurovascular cavernosa, PDE5 alone accounts for the breakdown of
process that can be initiated by recruitment of penile the majority (70%) of the formed cGMP (Morelli et al.
afferent signals (reflexogenic erection) and by visual, 2004). Relaxation of cavernous SMC increases blood
auditory, tactile, olfactory and imaginary stimuli (psy- flow into cavernosal sinuses and venous occlusion,
chogenic erection). Several brain regions have been resulting in penile engorgement and rigidity.
identified that are involved in the initiation of penile
erection. The effect of testosterone on these central
mechanisms is described in depth in Chapter 5.
12.3 Direct effects of testosterone
At the penile level, erection occurs when the two on penile growth and erection
sponge-like cavernous bodies (corpora cavernosa) Penile development from the urogenital sinus and
become engorged with blood. Corpora cavernosa are genital tubercle is a process which is not well under-
essentially composed of large endothelium-lined vas- stood, but two members of the Hedgehog (Hh) gene
cular lacunae separated by fibrous trabeculae. The family, Sonic hedgehog (Shh) and Indian hedgehog
amount of blood flow to the corpora cavernosa is (Ihh), expressed early during anogenital differenti-
regulated by the activity of SMC lining the penile ation, are known to direct division of the cloaca into
arteries and cavernous spaces in tight communication separate urogenital and anorectal sinuses. Shh/
with the overlying endothelial cells. Penile erection is mice have complete agenesis of the external genitalia
a vascular (hydraulic), three-step phenomenon and persistence of the cloaca; in humans, mutations
(increased arterial inflow, sinusoidal smooth muscle affecting Hedgehog signaling and its downstream
relaxation and decreased venous drainage). In the targets underlie several syndromes characterized by
flaccid penis, SMC of corpora cavernosa are con- anorectal malformation (Podlasek 2009). Androgens
tracted and penile blood inflow is low. During and a functioning AR are also necessary for normal
erection, relaxation of trabecular SMC results in development of the human penis. In humans, the penis
increased blood flow and pressure in the corpora grows in phases, initially during early gestation and
cavernosa and expansion of sinusoidal spaces. The then continuing until approximately the age of five.
expanded corpora cavernosa cause mechanical com- A latency period follows until puberty, when penile
pression of the emissary veins, restricting the venous size responds to the increase of testosterone levels.
outflow from the cavernosal spaces and facilitating an Growth ceases at the completion of pubertal growth
entrapment of blood in the cavernosal sinusoids. This despite continued high levels of circulating testoster-
blood engorgement finally results in penile rigidity. one. In a prospective, longitudinal population-based
A series of biochemical and hemodynamic events study of 728 Danish and 1234 Finnish infant boys it
that are associated with activation of the central was found that endogenous testosterone was

252
Chapter 12: Testosterone and erection

Fig. 12.1 Schematic representation of


(A) Sympathetic nerve the biochemical events leading to penile
flaccidity (A) or erection (B). The
Ca2+ enzymatic steps for which a role for
NA CH3 OH testosterone has been proposed are
CH3
represented. See plate section for color
VOC CLCAs version. (A) Noradrenalin (NA) binding
1 O generates inositol 1,4,5-trisphosphate
RhoA-GTP Testosterone (InsP3), which, by increasing intracellular
Smooth (active)
calcium (Ca2+) levels, activates Ca2+-
muscle cell GEFs GAPs sensitive chloride channels (CLCAs),
RhoA-GDP resulting in membrane depolarization
PLC
(inactive) with the diffusion of the stimulus to the
ROCK neighboring cells and the opening of
ATP voltage-operated channels (VOC). The
InsP3
ATP ATP increased Ca2+ flow promotes, through
calmodulin, activation of myosin light
CPI-17
chain (MLC) kinase (MLCK) and cell
P contraction. Cell contraction is also
Ca2+ MLCP obtained by altering the Ca2+ sensitivity
MLC MLC
SR through a NA-induced activation of a
MLCK second pathway, RhoA/ROCK, which
Ca2+/calmodulin complex
Activation increases, through a series of kinase
Inhibition activation, the sensitivity of MLC to Ca2+.
CONTRACTION Testosterone is supposed to negatively
regulate the latter event. (B) Nitric oxide
(NO) is generated by NO synthases in
(B) Testosterone either non-adrenergic, non-cholinergic
CH3
OH (NANC) neurons (nNOS) or endothelial
cells (eNOS). Both steps are positively
NANC nerves CH3
regulated by testosterone. Nitric oxide
eNOS Endothelial cell diffuses into SMC and activates a soluble
O
nNOS guanylate cyclase (sGC), which in turn
NO Ca2+
NO transforms GTP into cGMP. Cyclic GMP
activates protein kinase G (PKG), which,
through the indicated pathways, finally
decreases intracellular Ca2+ levels,
Smooth Hyperpolarization Ca2+-ATPase leading to relaxation. Phosphodiesterase
muscle cell NO K+ pump type 5 (PDE5) metabolizes cGMP into
GMP, thereby limiting its effects. The
latter event is positively controlled by
sGC cGMP PKG InsP3 testosterone. Other abbreviations: cGMP,
cyclic GMP; CPI-17, protein phosphatase
1 regulatory, subunit 14A; GAP, GTPase
GTP activating protein; GEF, guanine
Ca2+2+Ca2+ nucleotide exchange factor; MLCP,
GMP PDE5 Ca 2+
Ca2+ Ca myosin light chain phosphatase; PLC,
phospholipase C; rhoA, ras homolog
SR
CH3 OH
gene family member A; ROCK, rho-
CH3 MLCK P associated, coiled-coil containing protein
Activation MLC MLC kinase; SR, sarcoplasmic reticulum.
Inhibition MLCP
O
Testosterone
RELAXATION

significantly associated with penile size and growth rate Numerous studies in animal models have demon-
(Boas et al. 2006). The exact mechanism of penile growth strated a direct testosterone dependency of the erectile
cessation remains unresolved, but is probably not only process. Experimental studies in animals and human
due to downregulation of ARs (Levy et al. 1996; Baskin cell cultures indicate that testosterone controls, dir-
et al. 1997). Although Shh expression is partially andro- ectly or indirectly, the majority of mechanisms
gen dependent, in juvenile rats Shh signaling appears to leading to erection and detumescence. First of all, it
be independent of testosterone (Bond et al. 2010). controls the commitment of penile cells towards a

253
Chapter 12: Testosterone and erection

smooth muscle phenotype, favoring the functional Penson et al. 1996; 1997) and rabbit (Traish et al.
and structural integrity that is necessary for penile 1999) models of androgen ablation.
erection (Traish et al. 2005; Morelli et al. 2007; Yassin In addition, it has been demonstrated that testos-
and Saad 2008; Traish 2009; Corona and Maggi 2010). terone also regulates the activity of the RhoA/ROCK
There is, in fact, general consensus on a trophic effect pathway negatively, decreasing calcium sensitivity
of testosterone on penile architecture in different within penile SMC overall in both castration-induced
animal species such as rabbit (Traish et al. 1999; (Wingard et al. 2003) and diabetes-induced hypogo-
2003; Morelli et al. 2007), dog (Takahashi et al. nadism (Vignozzi et al. 2007). RhoA is a member of
1991), mouse (Palese et al. 2003) and rat (Shabsigh the small monomeric GTPase family which plays a
et al. 1998; Zhang et al. 1999; Shen et al. 2003). In role not only in mediating smooth muscle contrac-
castrated animals, elastic and smooth muscle fibers tion, through the activation of its downstream kinase
are replaced by collagenous ones, which may result in (ROCK), but also in the regulation of several cellular
fibrosis. It has also been shown that testosterone is processes, such as stress fiber formation and cell
involved in the maturation of penile tissue compos- migration.
ition by promoting the commitment of pluripotent Under the same experimental conditions (surgical
stem cells into the myogenic lineage, and inhibiting or chemical castration, diabetes/metabolic-syndrome-
their differentiation towards adipogenic lineage. induced hypogonadism), it has been shown that tes-
Androgen deficiency is associated with alterations in tosterone also positively regulates the expression of
dorsal nerve structure and endothelial morphology, PDE5 (Traish et al. 1999; Morelli et al. 2004; Zhang
and accumulation of adipocytes in the subtunical et al. 2005; 2006; Vignozzi et al. 2007; Filippi et al.
region of the corpora cavernosa, which could impede 2009). Because testosterone positively controls both
blood outflow during sexual stimulation and contrib- the enzymatic steps necessary for initiation (positive
ute to venous leak (Traish et al. 2005; Morelli et al. effect on NOS and negative on RhoA/ROCK) and the
2007; Yassin and Saad 2008). end (positive on PDE5) of the erectile process, its net
In addition, testosterone controls numerous effect on erection ends up as modest. Erections are
enzymatic activities within the corpora cavernosa. indeed still possible in hypogonadal conditions where
A significant role of testosterone has been demon- decreased cGMP formation, due to impaired NO
strated for regulating NO formation (acting on eNOS production, is most probably counterbalanced by
and/or nNOS) in several animal models. Low testos- reduced PDE5 activity and cGMP hydrolysis.
terone conditions include senescence (Garban et al. However, it should be considered that, both in
1995a; 1995b), diabetes mellitus type 1 and 2 (Vernet animals and men, testosterone acts mainly on sexual
et al. 1995), metabolic syndrome (Filippi et al. 2009), interest (Rochira et al. 2003; Vignozzi et al. 2005) and
adrenalectomy (Penson et al. 1997), hypophysectomy only partially on sexual potency. Accordingly, not all
(Penson et al. 1996) and castration (Mills et al. 1992; castrated men become impotent (Heim and Hursch
Chamness et al. 1995; Lugg et al. 1995; 1996; Penson 1979). Indeed, in prostatic cancer patients undergoing
et al. 1997; Park et al. 1999). In castrated rats, both medical or surgical castration, sexual intercourse was
testosterone (Chamness et al. 1995; Lugg et al. 1995; preserved in 2045% of subjects (Ellis and Grayhack
Park et al. 1999) and DHT (Lugg et al. 1995; Park 1963). Penile erections in response to powerful stim-
et al. 1999) replacement completely rescued NOS uli, such as erotic visual stimulation, have also been
activity. It is still rather unclear which of the two documented in hypo-androgenized individuals
major constitutive NOS isoforms, i.e. the neuronal (Kwan et al. 1983; Bancroft and Wu 1983; Greenstein
(nNOS) or endothelial (eNOS) isoform, is mainly et al. 1995). According to Davidson (1986), in hypo-
regulated by androgens in the rat penis. In the major- gonadal men the decreased libido apparently leads to
ity of studies androgens increased nNOS protein a secondary decrease in erectile performance during
(Chamness et al. 1995; Park et al. 1999; Marin et al. sexual situations, due probably to lack of interest or
1999) or mRNA (Reilly et al. 1997; Schirar et al. 1997; pleasure. Hence, erections are still possible without
Park et al. 1999); although positive effects on eNOS androgens, although less frequent because of low
were also reported (Marin et al. 1999; Filippi et al. sexual interest. It has been reported since ancient
2009). Other studies were negative on eNOS (Park times that eunuchs who were castrated after puberty
et al. 1999) or nNOS in both rat (Lugg et al. 1996; were capable of maintaining erections. Accordingly, it

254
Chapter 12: Testosterone and erection

was a custom in ancient Rome that women would use graphical plot of the sensitivity, or true positive rate,
more potent eunuchs for pleasure without the risk of vs. false positive rate (1  specificity or 1  true
procreation (Dettenhofer 2009). Infants frequently negative rate), for a binary classifier system as its
have erections upon the slightest tactile stimulation, discrimination threshold is varied. The ROC curve
but, until puberty, boys do not engage in sexual activ- analysis for severely impaired PSV (<25 cm/s) showed
ity outside the context of play. The paradox of a an overall accuracy of 65.3% in predicting low testos-
normal erection (upon adequate sexual stimulation) terone (p < 0.0001), with a specificity and sensitivity to
in hypo-androgenized individuals, in which NO for- detect abnormal testosterone (below 12 nmol/l) of
mation is substantially hampered, finds an intriguing 72.5% and 51.6%, respectively (see Fig. 12.2B).
solution in the observation published almost three Interestingly, in patients with ED, the evaluation
decades ago by Kwan et al. (1983). They found that of penile flow could be used not only to identify those
the erectile response to explicit pornographic films in deserving further investigation, but also to stratify
hypogonadal individuals was comparable to that of future cardiovascular risk (Corona et al. 2011b). It
normal ones, while the time to penile detumescence has been shown that, in ED subjects, a dynamic PSV
was prolonged by a factor of two in the hypogonadal below 25 cm/s is associated with a relevant increase of
men as compared to eugonadal controls. The demon- cardiovascular risk (Corona et al. 2010b): the risk of
stration that cGMP breakdown through PDE5 is major adverse cardiovascular events (MACE)
androgen dependent helps in understanding this increases by 5% for each 5 cm/s decrement of the
apparent paradox, because, in a low-androgen milieu, dynamic PSV. In particular, male hypogonadism has
cGMP catabolism is impaired (see Fig. 12.1; Morelli been associated with increased mortality in patients
et al. 2004; Zhang et al. 2005). affected by specific diseases, such as hypopituitarism,
Vasoactive intestinal peptide (VIP) is one of the Klinefelter syndrome, mental retardation, and in spe-
first penile neurotransmitters characterized for facili- cific populations, such as veterans and Japanese men,
tating erectile function. It signals though the G-pro- with at least one cardiovascular factor (Corona et al.
tein-coupled receptor VPAC2, and, upon Gs a and 2011b). In line with these findings, it has been
adenylyl cyclase activation, it stimulates cAMP for- recently reported that, in ED subjects, low testoster-
mation. Within the penis of hypogonadal rats, VIP one predicted MACE lethality (Corona et al. 2010c)
signaling was substantially increased (Zhang et al. and forthcoming MACE; the latter particularly in
2011), partially rescuing castration-induced ED. Tes- normal weight and overweight subjects (Corona
tosterone supplementation to hypogonadal rats par- et al. 2011c).
tially downregulated this pathway. Penile nocturnal erections are a naturally occur-
ring phenomenon during rapid eye movement (REM)
sleep in all normal healthy males from infancy to old
12.4 Association between androgen age, and they have no association with dream content
level and erectile activity in men with (Montorsi and Oettel 2005). Although it is well
accepted that sleep-related erections (SREs) deterior-
erectile dysfunction ate with ED, quite surprisingly the physiological role
The most widely used parameter to predict adequacy of sleep-related erections is still poorly understood.
of penile circulation is cavernous peak systolic vel- Testosterone seems to play a crucial role in the regu-
ocity (PSV, cm/s), measured 520 minutes after an lation of nocturnal erections. An androgenic control
intracavernous injection of a vasodilating agent of SREs during REM sleep has been postulated for a
(Corona et al. 2008). Fig. 12.2 shows an association long time. Karacan and Hirshkowitz (1991) noted
between circulating testosterone and penile blood that, in boys, SREs increase at the onset of puberty,
flow, as well as reported erectile activity, in a large suggesting a role of testosterone in such a process. In
series of subjects attending an andrological clinic for addition, testosterone levels peak near the transition
sexual dysfunction. As shown in Fig. 12.2A, penile from non-REM to REM sleep close to the onset of an
blood flow, as detected by penile color Doppler ultra- SRE episode (Roffwarg et al. 1982). In the early 1980s
sound (PCDU) after PGE1 injection (10 mg), is posi- Cunningham et al. (1982) reported that hyperprolac-
tively related to testosterone plasma levels. The tinemia-induced hypogonadism was associated with a
receiver operating characteristic (ROC) curve is a reduction of SREs. These results were confirmed by

255
Chapter 12: Testosterone and erection

ROC of T for dynamic PSV<25 cm/s


(A) (B) 1.0
18 Total T=12 nmol/l
Adj. r = 0.090;
0.8
p <0.0001, n = 2048
Total testosterone (nmol/l)

17

Sensitivity
0.6

16
0.4

15
0.2

14 0.0
0.0 0.2 0.4 0.6 0.8 1.0
I II III IV
Dynamic PSV quartiles 1-specificity

(C) 18 (D) 18

Total testosterone (nmol/l)


Total testosterone (nmol/l)

17 17

16 16

15 15

14 Adj. r = -0.093; 14 Adj. r = -0.048;


p <0.0001, n = 3527 p = 0.004, n = 3574
13 13

<25 2550 5075 >75%


No Mild Moderate Severe
Perceived reduced nocturnal erections Erection absent
Fig. 12.2 Age-adjusted association (Adj. r) of reported erectile activity with blood levels of total testosterone (T) in the indicated number (n) of
subjects with sexual dysfunction, studied at the University of Florence (Italy). (A) Association of maximal peak systolic velocity (PSV) at PCDU
(after 10 mg PGE1), reported as quartiles, and total T. (B) ROC curve of PSV < 25 cm/s for total testosterone in the same population as in panel
A. The arrow shows the sensitivity (ordinate) and 1 specificity (abscissa) for total testosterone = 12 nmol/l. (C) Association between perceived
reduced nocturnal/morning erections of increasing intensity (no, mild, moderate, severe) and total T. (D) Association between reported severe
ED (erection absence, percentage of cases) and total T.

Kwan et al. (1983) around the same time and more SREs can be easily measured non-invasively
recently by Carani et al. (1995). Testosterone levels through the use of a nocturnal penile tumescence
correlate with the measured frequency of sleep (NPT) test. Interestingly, the decreased frequency of
erections (Cunningham et al. 1982; Roffwarg et al. SREs has been recognized recently as a possible pre-
1982; Kwan et al. 1983; Karacan and Hirshkowitz dictive symptom of testosterone deficiency in
1991; Carani et al. 1995; Hirshkowitz and Schmidt community-dwelling men (Wu et al. 2010). It has been
2005; Montorsi and Oettel 2005; Rochira et al. 2006), reported recently that investigation of SREs, through a
and TRT in hypogonadal men has been shown to validated (and costless) questionnaire (SIEDY item
increase the frequency, magnitude, duration and no. 13, Petrone et al. 2003), is able to predict NPT
rigidity of SREs (Cunningham et al. 1982; OCarroll test results with an accuracy of about 70%. Using
et al. 1985; Hirshkowitz and Schmidt 2005; Montorsi this questionnaire, it has been demonstrated that per-
and Oettel 2005; Rochira et al. 2006). ceived reduced SREs were significantly associated

256
Chapter 12: Testosterone and erection

with total (Fig. 12.2C), analog free, calculated free and testosterone (testosterone concentration < 12 nmol/l
calculated bioavailable testosterone, even after adjust- or <346 ng/dl). Interestingly, meta-regression analy-
ment for age (Corona et al. 2011d). sis demonstrated that the effect of TRT on erectile
In contrast to the clear-cut androgen dependence function was inversely related to the baseline testos-
of both penile blood flow and spontaneous/nocturnal terone concentration. Hence, the more severe the
erections, reported sex-related erections were less hypogonadism, the more significant or impressive
clearly associated with testosterone blood levels. Only are the results obtained with TRT. Conversely, no
subjects reporting a severe ED show lower blood effect was observed for baseline testosterone levels
levels of testosterone (Corona and Maggi 2010). This higher than 12 nmol/l.
indicates that penile erection is associated with the A subsequent meta-analysis (Boloa et al. 2007),
androgen milieu, even though other factors (i.e. including only trials devoted to men with sexual
intrapsychic, relational) might mitigate the patients dysfunction at baseline, emphasized the possible role
perception during sexual activity. Accordingly, Rho- of aging as a moderator in evaluating the effect of
den et al. (2002), in a large consecutive series of 1071 TRT on sexual function. In particular, analyzing
mainly eugonadal men aged from 40 to 90 years, the studies according to mens age, they reported a
found no significant association between serum tes- sizable and significant effect of TRT on erectile func-
tosterone levels and the prevalence or severity of ED tion in the two trials including young patients, and a
as assessed by the questionnaire of the simplified minimal and non-significant effect in those including
International Index of Erectile Function (IIEF-5). older ones (mean age > 50 years). This last observa-
tion is not surprising. In fact, while hypogonadism
12.5 Effects of testosterone therapy can be the main cause of ED in younger patients, it is
generally only one element of a multifactorial ED in
on erection older ones.
Since the early beginnings of testosterone therapy of
hypogonadal patients it has been known that testos-
terone restores normal male sexual behavior and
12.6 Combined therapy with
erectile function (see Chapter 5). However, studies testosterone and phosphodiesterase
on the effect of TRT are surprisingly scanty, of short type 5 inhibitors in patients with
duration and often not placebo controlled. In order to
emphasize the most important outcomes of TRT, erectile dysfunction
meta-analysis was performed. In fact meta-analysis Oral therapy with inhibitors of the phosphodiesterase
can pool trials with different results and offer clin- type 5 (PDE5i), e.g. sildenafil, vardenafil and tadalafil,
icians and patients a single best estimate of the effect is highly effective in treating ED (Tsertsvadze et al.
of TRT. Meta-analysis is particularly useful when 2009). However, in placebo-controlled phase III clin-
there are a variety of reports with low statistical ical trials and post-marketing evaluation, approxi-
power; thus, pooling data can improve power and mately 15 to 40% of patients do not respond to this
provide more robust results. medication (Tsertsvadze et al. 2009). Because PDE5 is
The first meta-analysis on TRT for ED in hypo- androgen dependent (Morelli et al. 2004), it has been
gonadal patients confirmed the significant improve- suggested that the combination of TRT and PDE5
ment of erections after initiation of testosterone inhibitors can be used to improve PDE5i outcomes
therapy (Jain et al. 2000). Pooled data on placebo- in hypogonadal men. All observations emphasize that
controlled studies showed an improvement of erectile hypogonadism must be ruled out and, if present,
function in 36 of 55 men treated with testosterone; adequately treated, before the prescription of any
whereas significantly fewer men responded to placebo PDE5i. On the other hand, in an uncontrolled study
treatment (9 out of 45) (Jain et al. 2000). A meta- Greenstein et al. (2005) reported that the combination
analysis of 17 randomized placebo-controlled clinical of sildenafil was able to restore the 100% success rate
trials (Isidori et al. 2005) indicated that, in compari- in 48 mild hypogonadal ED patients, previously
son to placebo, a significant, but moderate, improve- treated only with testosterone gel.
ment of all aspects of sexual function was found in the At present, various uncontrolled studies (Kalinch-
studies of middle-age and elderly men with low enko et al. 2003; Chatterjee et al. 2004; Foresta et al.

257
Chapter 12: Testosterone and erection

Table 12.1 Trials assessing the effect of combined therapy with testosterone (T) and sildenafil/tadalafil in men with erectile dysfunction
unresponsive to testosterone (11), to PDE5i (13,6,10,12) or directly the combination of the two treatments (4,5,7,8, 9,13)

References n Baseline testosterone Therapy Overall efficacy (%)


levels (ng/dl)
1 Aversa et al. 2003a 20 <400 T patch/sildenafil 100 mg 80
2 Kalichenko et al. 2003 120 <340 Oral TU/sildenafil 100 mg 70
3 Shabsigh et al. 2004 a
75 <400 T gel/sildenafil 70
100 mg
4 Chatterjee et al. 2004 12 <400 (75%) T im/sildenafil 100
50100 mg
5 Foresta et al. 2004 15 <200 T patch/sildenafil 50 mg Normal NPT test par
6 Shamloul et al. 2005 40 <340 Oral TU/sildenafil Improvement
50100 mg
7 Greenstein et al. 2005 31 <400 T gel/sildenafil 63
100 mg
8 Tas et al. 2006 23 <400 T im/sildenafil 34
50100 mg
9 Rochira et al. 2006a 24 < 200 T im/sildenafil Improved NPT test par
50 mg
10 Hwang et al. 2006 32 <300 Oral TU/sildenafil 100 mg 57
11 Rosenthal et al. 2006 90 <350 T gel/sildenafil 92
100 mg
12 Yassin et al. 2006 69 <340 T gel/tadalafil 65
20 mg
13 Buvat et al. 2011a 73 <300 T gel/tadalafil 51
10 mg daily
Abbreviations: TU, testosterone undecanoate; NPT, nocturnal penile tumescence; Par, parameters.
a
Placebo-controlled studies.

2004; Shamloul et al. 2005; Hwang et al. 2006; PDE5i responsiveness. The first study included 20
Rosenthal et al. 2006; Tas et al. 2006; Yassin et al. patients with arteriogenic ED as diagnosed by
2006) and four randomized, placebo-controlled dynamic color duplex ultrasound (PCDU). These
studies (Aversa et al. 2003; Shabsigh et al. 2004; men had normal sexual desire, serum levels of total
Rochira et al. 2006; Buvat et al. 2011) suggest that and free testosterone in the lower quartile of the
hypogonadism might hinder the effects of PDE5i on normal range, and had not responded to the highest
erectile function (Table 12.1). In particular, the com- dose of sildenafil (100 mg) on six consecutive
bination of testosterone and PDE5i is able to improve attempts (Aversa et al. 2003). Patients were random-
the overall efficacy from 34 to 100% (see Table 12.1). ized to transdermal non-scrotal testosterone patches
In addition, one study suggested that the proportion (5 mg/d; n = 10) or placebo patches (n = 10), and
of patients improved by the combination with testos- received 100 mg sildenafil tablets on demand. The
terone increased with the duration of testosterone PCDU revealed a significant increase of arterial
(65% after 10 weeks vs. 43% after only 4 weeks) inflow to cavernous arteries of the penis in the testos-
(Aversa et al. 2003). terone-treated men; whereas this parameter remained
Three randomized, controlled studies support the unchanged in the placebo group. Effects on erectile
concept of a positive interaction between TRT and function were assessed by the International Index of

258
Chapter 12: Testosterone and erection

Erectile Function (IIEF) questionnaire (Rosen et al. additional effect of testosterone administration to
1997). Compared to placebo plus sildenafil, treatment men optimally treated with PDE5i was encountered.
with testosterone and sildenafil resulted in a signifi- However, the differences between the testosterone
cantly increased score of erectile function domain, the and placebo groups were significant for both Erectile
intercourse satisfaction domain, and the overall satis- Function Domain Score of the IIEF and Sexual
faction domain of the IIEF. The scores of the sexual Encounter Profile 3 in those men with baseline tes-
desire domain and orgasmic function domain tosterone  10.4 nmol/l.
remained unchanged, indicating that the treatment These three randomized, placebo-controlled stud-
effect of testosterone was not only due to central ies provide evidence that patients with ED and low-
effects on sexual desire. The Global Assessment Ques- normal or subnormal testosterone levels, who do not
tion (GAQ) Has the treatment you received . . . respond to PDE5i therapy, might benefit from a com-
improved your erections was positively affirmed by bined therapy. However, the real efficacy of this
80% of men in the testosterone/sildenafil group com- approach needs to be proven in larger placebo-con-
pared to 10% in the placebo/sildenafil group at the trolled studies, because the meta-analysis by Tserts-
end of treatment (Aversa et al. 2003). vadze et al. (2009) did not support this view. It should
This pilot study was followed by a randomized, be noted that the recent study by Buvat was not
double-blind, placebo-controlled, 12-week multicen- included in the Tsertsvadze meta-analysis.
ter study in 70 men with low or low-normal serum Nevertheless, an international consultation, col-
testosterone (morning levels before 10 a.m. < 13.88 laborating with major urological and sexual medicine
nmol/l) and non-responders to 100 mg of sildenafil societies convened in Paris, July 2009 (International
during a 4-week run-in period (Shabsigh et al. 2004). Consultation of Sexual Medicine, ICSM-5), and rec-
Patients were randomly assigned to therapy with pla- ommended screening for testosterone deficiency in all
cebo gel and sildenafil (group I, n = 33) or 5 g/d of patients with ED (Buvat et al. 2010).
testosterone gel and sildenafil (group II, n = 37).
While the severity of ED was similar in both groups
at baseline, the erectile function domain, as well as the 12.7 Prevalence of testosterone
orgasmic function domain of the IIEF, improved sig- deficiency in patients with erectile
nificantly in group II at study week four, and scores
remained at this level up to the end of the study. dysfunction
However, it should be noted that the difference Recognition of hypogonadism in subjects with sexual
between both groups lost significance following week dysfunction has been greatly helped by recent consen-
four because of improvement of the total IIEF score sus among professional societies (Petak et al. 2002;
and score of the erectile function domain in group I. In Wang et al. 2009; Bhasin et al. 2010). It has been
addition, no control group treated only with testoster- universally recognized that diagnosis of androgen
one and oral placebo was included in this study. deficiency should be made only in symptomatic men
Recently, results from a multicenter, multi- with unequivocally low serum testosterone levels, as
national, double-blind, placebo-controlled study of neither criterion is consistently reliable when con-
173 men, 4580 years of age, non-responders to treat- sidered alone. All the guidelines recommend that total
ment with different PDE5 inhibitors, with baseline testosterone, the major parameter to be measured,
total testosterone levels  13.88 nmol/l or bioavailable should be sampled in the morning on at least two
testosterone  3.47 nmol/l were reported (Buvat et al. separate occasions before making a diagnosis. Testos-
2011). Men were first treated with tadalafil 10 mg terone substitution should be offered to symptomatic
once a day for 4 weeks; if not successful, they were individuals when circulating total testosterone is
randomized to receive placebo or 5 mg of testosterone below 8 nmol/l (231 ng/dl) (Wang et al. 2009). In
gel, to be increased to 10 mg testosterone if results addition, there is also general agreement that a total
were clinically unsatisfactory. Erectile function testosterone level above 12 nmol/l (346 ng/dl) does
improved progressively over a period of at least 12 not require substitution. When total testosterone is
weeks in both the placebo and testosterone treatment between 8 and 12 nmol/l in the presence of typical
groups. In the overall population, with a mean base- hypogonadal symptoms, a testosterone treatment trial
line testosterone level of 11.7  3.47 nmol/l, no should be considered (Wang et al. 2009).

259
Chapter 12: Testosterone and erection

Among the different symptoms, sexual dysfunc-

Prevalence of hypogonadism (%)


30
tion represents the most specific symptom associated
25
with low testosterone (Wu et al. 2010). Recent data
from the EMAS recognized that a triad of sexual 20
symptoms (low libido, and reduced spontaneous
15
and sex-related erections) is the only syndromic
association with decreased testosterone levels (Wu 10
et al. 2010); whereas other proposed hypogonadal
5
symptoms did not segregate with the androgen defi-
ciency. In that large European survey, the simultan- 0
< 8 nmol/l < 10.4 nmol/l < 12 nmol/l < 11 nmol/l
eous presence of the three sexual symptoms + 3 sexual
combined with a total testosterone level of less than + 1 sexual symptom symptoms
11 nmol/l and a free testosterone level of less than Fig. 12.3 Prevalence (%) of hypogonadism in subjects with sexual
220 pmol/l were therefore considered the minimal dysfunction (n = 3608), studied at the University of Florence (Italy),
criteria for the diagnosis of hypogonadism (Wu et al. according to different biochemical thresholds for low testosterone.
The proportion of hypogonadal subjects as derived from the new
2010). Conversely, in subjects with total testosterone proposed criteria based on the EMAS (Wu et al. 2010) is also
below 8 nmol/l, the presence of sexual symptoms was represented (three sexual symptoms and total testosterone
unhelpful. below 11 nmol/l).
In epidemiological studies in ED subjects, serum
testosterone below 10.4 nmol/l was found in 12% of medicine, as no study included a control group of
7000 men, compiled from nine large series (including age-matched men without ED.
4% of 944 men less than 50 years old, and 14.7% of Fig. 12.3 shows the different prevalences of hypo-
4342 men more than 50 years old) (Buvat and Bou gonadism in 3608 subjects referred to an andrological
Jaoud 2006). In one consecutive series of 1022 men clinic for sexual dysfunction (University of Florence).
with ED, Buvat and Lemaire (1997) detected serum All subjects were, by definition, symptomatic, because
concentrations of testosterone below 10.4 nmol/l in they reported at least one symptom of sexual dysfunc-
8.0% of men. However, 40% of these patients had tion. The data are essentially in keeping with results
normal serum levels at repeated determination (Buvat from Bodie et al. (2003) and Khler et al. (2008), and
and Lemaire 1997). Pituitary tumors were discovered show variable detection of male hypogonadism
in two men with low testosterone. Determination of ranging from 7.4% (below 8 nmol/l) to 27.2% (below
testosterone only in cases of low sexual desire or 12 nmol/l). However, when the strict criteria from the
abnormal physical examination would have over- EMAS were applied (total testosterone below 11
looked 40% of men with low testosterone, and 37% nmol/l and at least three sexual symptoms), the
of men subsequently improving during testosterone resulting percentage of ED men with hypogonadism
substitution therapy (Buvat and Lemaire 1997). was lower (8.2%), but still higher than in the general
A larger study involving 3547 men with ED revealed population, according to the EMAS (2.1%).
a prevalence of testosterone deficiency (serum con-
centration less than 9.7 nmol/l) of 18.7% (Bodie et al.
2003). In another consecutive series of 2794 men with 12.8 Effects of sexual activity and
ED aged 2580 years, testosterone was <6.94 nmol/l treatment of erectile dysfunction
in 7%, <10.4 nmol/l in 23%, <12 nmol/l in 33%, and
<13.88 nmol/l in 47% of men (Khler et al. 2008). on testosterone
The difference in the prevalence of testosterone Studies in rhesus macaques indicate that testosterone
deficiency in patients with ED could be explained by plasma levels are increased by exposure to a receptive
different patient populations, different age of men female and copulation (Herndon et al. 1981). Hence,
with ED, differences in primary, secondary or tertiary sexual activity and sex hormones appear to be intim-
care centers, different definitions of low testosterone ately related. Accordingly, Fabbri et al. (1988)
levels, or single versus repeated testosterone deter- reported that, in non-organic impotent men, both
minations. It should be noted that none of these bioactive LH and testosterone serum levels were sig-
studies really fulfils the principles of evidence-based nificantly lower than in normal men. However, it is

260
Chapter 12: Testosterone and erection

still unclear whether, in mating male individuals, tes- higher sexual intercourse frequency, have a lower
tosterone is higher to allow better sexual and repro- prevalence of biochemical and clinical hypogonadism
ductive fitness (affecting libido/penile erections and/ (Fisher et al. 2009). Similar results were previously
or spermatogenesis) or the reverse is true: sexual reported by van Anders et al. (2007). In Coronas
activity positively affects testosterone production. An study, the sexual-frequency-dependent reduction of
often-cited, single observation, published in Nature both total and free testosterone was not accompanied
almost 40 years ago, opted for the second scenario. by a compensatory rise in LH levels, suggesting the
An island resident observed an increase in beard presence of a central, hypothalamic-pituitary failure.
growth on the day preceding, and during, his occa- Similar results were obtained in a rat experimental
sional visits to his mainland lover (Anonymous 1970). model (Vignozzi et al. 2009). Hypogonadotropic
During the following years only scanty reports hypogonadism was observed occurring three months
substantiated this anecdotal account, demonstrating after bilateral denervation of the penis, obtained
a time-related rise in testosterone level during sexual through resection of the rats cavernous nerves (Vig-
intercourse (Fox et al. 1972; Dabbs and Mohammed nozzi et al. 2009). It was speculated that sexual
1992) or exposure to erotic movies (LaFerla et al. inactivity, induced by cavernous denervation, could
1978). Conversely, other reports addressing the ques- lead to a state of overt hypogonadotropic hypogonad-
tion of a sexual-activity-induced rise in testosterone ism (Vignozzi et al. 2009). This could also be the case
plasma levels were negative (Raboch and Starka 1973; in patients, considering that decreased penile blood
Stearns et al. 1973; Lee et al. 1974; Kraemer et al. flow and decreased sexual activity were associated
1976). However, Stoleru et al. (1993), sampling male with lower androgens and presence of hypogonadal
volunteers every 10 min for 12 h, found an increased symptoms, with inappropriately low LH levels
pulsatile release of LH, with a concomitant increase in (Corona et al. 2009).
testosterone, occurring soon after exposure to erotic, Jannini et al. produced evidence that substantiated
but not neutral, movies. In normal healthy men and the hypothesis of an LH-mediated, sex-induced drive
patients with ED, serum levels of testosterone increase in testosterone production (Jannini et al. 1999; Carosa
significantly during the tumescence, as well as the et al. 2002; 2004). In a controlled, non-randomized
rigidity phase of penile erection, and return to baseline study they demonstrated that effective psychological,
in the detumescence phase (Becker et al. 2000; 2001). medical (prostaglandin E1, yohimbine) or mechanical
In a consecutive series of 2302 male patients with (vascular surgery, penile prostheses, vacuum devices)
ED (mean age 53.2  12.5 years) an association was therapy of ED leads to a sustained increase of serum
found between SIEDY Scale 2 score (which measures testosterone levels (Jannini et al. 1999). The testoster-
the relational component of ED: Petrone et al. 2003) one rise they found was independent of the kind of
and decreased intercourse frequency, severe ED, therapies employed, but strictly related to the success-
lower dynamic PSV at PCDU, and clinical and bio- ful outcome of therapeutic intervention. Hence, they
chemical hypogonadism (Corona et al. 2009). Alter- speculated that sexual inertia resets the reproductive
native models were explored using these different axis to lower activity, somehow inducing a secondary
factors as dependent variables, in order to evaluate hypogonadism, characterized by reduced LH bioac-
the specific relationship among the parameters. Mul- tivity (Carosa et al. 2002). Consequently, restoring sex
tiple logistic regression analysis indicated that low normalizes sex hormones, including bioavailable LH
penile blood flow and decreased intercourse fre- and testosterone (Jannini et al. 1999; Carosa et al.
quency are bi-directionally coupled to poor relational 2004). However, RCTs are awaited to prove this inter-
domain, while the association with hypogonadism esting hypothesis.
was mediated through sexual hypo-activity or inertia.
In other words, this suggests that good sex is the
prerequisite for a good couple relationship and a good 12.9 Conclusion
couple relationship is the cornerstone for successful There is clear evidence from experimental studies that
and happy couple sexual activity, which is accompan- testosterone influences erectile function not only
ied by higher testosterone levels (Corona et al. 2009). indirectly by increased libido, but has direct effects
Accordingly, it was described that among subjects on the penis. Whereas testosterone substitution ther-
with sexual dysfunction, unfaithful men, reporting apy is effective for treatment of ED in hypogonadal

261
Chapter 12: Testosterone and erection

patients, the effects of testosterone on erectile func-


tion in normal men seem to be marginal. Recent erectile function. Testosterone not only enhances
spontaneous sleep-related erections, but to a
studies suggest that therapy combining testosterone
lesser degree also erectile response.
and phosphodiesterase type 5 inhibitors could be
 In eugonadal men, variations of testosterone
useful in so-called PDE5i non-responders with levels within the normal range or levels
low-normal or subnormal testosterone levels. exceeding the upper limit of normal have no or
very limited influence on erectile function.
12.10 Key messages  The true prevalence of testosterone deficiency as
 Positive effects of testosterone on erection are a cause for ED is not known, but seems to be less
mediated by central stimulation of libido and than 20%. Recent consensus statements
sexual activity, but also by direct effects on the recommend screening for testosterone
penis. deficiency in patients with ED.
 Experimental studies suggest that the integrity of  Randomized, placebo-controlled studies indicate
the smooth muscles of the penile arteries and the that patients with ED and low-normal or
corpora cavernosa, as well as the biological activity subnormal testosterone levels benefit from
of nitric oxide, the predominant cellular transmitter therapy combining testosterone and
for normal erection, are androgen dependent. phosphodiesterase type 5 inhibitors.
 Impaired erectile function is a classical symptom  Preliminary, non-randomized studies suggest
of hypogonadism. Testosterone therapy of that effective therapy of ED increases serum
hypogonadal patients significantly improves testosterone levels.

12.11 References Jonas U (2001) Cavernous and based approach. J Urol 169:
systemic testosterone plasma levels 22622264
Anonymous (1970) Effects of sexual during different penile conditions in
activity on beard growth in man. Boloa ER, Uraga MV, Haddad RM,
healthy males and patients with
Nature 226:869870 erectile dysfunction. Urology
Tracz MJ, Sideras K, Kennedy CC,
Caples SM, Erwin PJ, Montori VM
Aversa A, Isidori AM, Spera G, Lenzi 58:435440
(2007) Testosterone use in men with
A, Fabbri A (2003) Androgens Berridge MJ (2008) Smooth muscle cell sexual dysfunction: a systematic
improve cavernous vasodilation and calcium activation mechanisms. review and meta-analysis of
response to sildenafil in patients J Physiol 586:50475061 randomized placebo-controlled
with erectile dysfunction. Clin
Bhasin S, Cunningham GR, Hayes FJ, trials. Mayo Clin Proc 82:2028
Endocrinol (Oxf) 58:632638
Matsumoto AM, Snyder PJ, Bond CW, Angeloni NL, Podlasek CA
Bancroft J, Wu FC (1983) Changes in Swerdloff RS, Montori VM; Task
erectile responsiveness during (2010) Analysis of testosterone
Force, Endocrine Society (2010) effects on sonic hedgehog signaling
androgen replacement therapy. Testosterone therapy in men with
Arch Sex Behav 12:5966 in juvenile, adolescent and adult
androgen deficiency syndromes: an sprague dawley rat penis. J Sex Med
Baskin LS, Sutherland RS, DiSandro Endocrine Society clinical practice 7:11161125
MJ, Hayward SW, Lipschutz J, guideline. J Clin Endocrinol Metab
Cunha GR (1997) The effect of 95:25362559 Buvat J, Bou Jaoud G (2006)
testosterone on androgen receptors Significance of hypogonadism in
Boas M, Boisen KA, Virtanen HE, erectile dysfunction: World J Urol
and human penile growth. J Urol Kaleva M, Suomi AM, Schmidt IM,
158:11131118 24:657667
Damgaard IN, Kai CM, Chellakooty
Becker AJ, Uckert S, Stief CG, Truss M, Skakkebaek NE, Toppari J, Main Buvat J, Lemaire A (1997) Endocrine
MC, Machtens S, Scheller F, Knapp KM (2006) Postnatal penile length screening in 1,022 men with erectile
WH, Hartmann U, Jonas U (2000) and growth rate correlate to serum dysfunction: clinical significance
Cavernous and systemic testosterone levels: a longitudinal and cost-effective strategy. J Urol
testosterone levels in different study of 1962 normal boys. Eur 158:17641767
phases of human penile erection. J Endocrinol 154:125129 Buvat J, Maggi M, Gooren L, Guay AT,
Urology 56:125129 Bodie J, Lewis J, Schow D, Monga M Kaufman J, Morgentaler A,
Becker AJ, Uckert S, Stief CG, Scheller (2003) Laboratory evaluations of Schulman C, Tan HM, Torres LO,
F, Knapp WH, Hartmann U, erectile dysfunction: an evidence Yassin A, Zitzmann M (2010)

262
Chapter 12: Testosterone and erection

Endocrine aspects of male sexual Corona G, Mannucci E, Mansani R, Corona G, Rastrelli G, Ricca V, Maggi
dysfunctions. J Sex Med 7: Petrone L, Giommi R, Mancini M, M (2011a) Testosterone deficiency
16271656 Bartolini M, Forti G, Maggi M in the aging male and its
Buvat J, Montorsi F, Maggi M, Porst H, (2004) Aging and pathogenesis of relationship with sexual
Kaipia A, Colson MH, Cuzin B, erectile dysfunction. Int J Impot Res dysfunction and cardiovascular
Moncada I, Martin-Morales A, 16:395402 diseases. Horm Mol Biol Clin
Yassin A, Meuleman E, Eardley I, Corona G, Fagioli G, Mannucci E, Investig 4:509520. DOI 10.1515/
Dean JD, Shabsigh R (2011) Romeo A, Rossi M, Lotti F, Sforza hmbci.2010.048
Hypogonadal men nonresponders A, Morittu S, Chiarini V, Casella G, Corona G, Rastrelli G, Vignozzi L,
to the PDE5 inhibitor tadalafil Di Pasquale G, Bandini E, Forti G, Mannucci E, Maggi M (2011b)
benefit from normalization of Maggi M (2008) Penile Doppler Testosterone, cardiovascular disease
testosterone levels with a 1% ultrasound in patients with ED: role and the metabolic syndrome. Best
hydroalcoholic testosterone gel in of peak systolic velocity measured in Pract Res Clin Endocrinol Metab
the treatment of erectile dysfunction the flaccid state in predicting 25:337353
(TADTEST study). J Sex Med arteriogenic ED and silent coronary Corona G, Rastrelli G, Monami M,
8:284293 artery disease. J Sex Med 5: Melani C, Balzi D, Sforza A, Forti G,
Carani C, Granata ARM, Bancroft J, 26232634 Mannucci E, Maggi M (2011c) Body
Marrama P (1995) The effects of Corona G, Mannucci E, Lotti F, Boddi mass index regulates
testosterone replacement on V, Jannini EA, Fisher AD, Monami hypogonadism-associated CV risk:
nocturnal penile tumescence and M, Sforza A, Forti G, Maggi M results from a cohort of subjects
rigidity and erectile response to (2009) Impairment of couple with erectile dysfunction. J Sex Med
visual erotic stimuli in hypogonadal relationship in male patients with 8:20982105
men. Psychoneuroendocrinology sexual dysfunction is associated
Corona G, Rastrelli G, Balercia G,
20:743753 with overt hypogonadism. J Sex Med
Sforza A, Forti G, Mannucci E,
6:25912600
Carosa E, Benvenga S, Trimarchi F, Maggi M (2011d) Perceived reduced
Lenzi A, Pepe M, Simonelli C, Corona G, Lee DM, Forti G, OConnor sleep-related erections in subjects
Jannini EA (2002) Sexual inactivity DB, Maggi M, ONeill TW, with erectile dysfunction:
results in reversible reduction of LH Pendleton N, Bartfai G, Boonen S, psychobiological correlates. J Sex
bioavailability. Int J Impot Res Casanueva FF, Finn JD, Giwercman Med 8:17801788
14:9399 A, Han TS, Huhtaniemi IT, Kula K,
Cunningham GR, Kracan I, Ware JC,
Lean ME, Punab M, Silman AJ,
Carosa E, Martini P, Brandetti F, Di Lantz GD, Thornby JI (1982) The
Vanderschueren D, Wu FC; EMAS
Stasi SM, Lombardo F, Lenzi A, relationships between serum
Study Group (2010a) Age related
Jannini EA (2004) Type testosterone and prolactin levels
changes in general and sexual health
V phosphodiesterase inhibitor and nocturnal penile tumescence
in middle-aged and older men:
treatments for erectile dysfunction (NPT) in impotent men. J Androl
results from the European Male
increase testosterone levels. Clin 3:241247
Ageing Study (EMAS). J Sex Med
Endocrinol (Oxf) 61:382386
7:13621380 Dabbs JM Jr, Mohammed S (1992)
Chamness SL, Ricker DD, Crone JK, Corona G, Monami M, Boddi V, Male and female salivary
Dembeck CL, Maguire MP, Burnett Cameron-Smith M, Lotti F, de Vita testosterone concentrations before
AL, Chang TS (1995) The effect of G, Melani C, Balzi D, Sforza A, Forti and after sexual activity. Physiol
androgen on nitric oxide synthase G, Mannucci E, Maggi M (2010b) Behav 52:195197
in the male reproductive tract of the Male sexuality and cardiovascular Davidson JM (1986) Androgen
rat. Fertil Steril 63:11011107 risk. A cohort study in patients with replacement therapy in a wider
Chatterjee R, Wood S, McGarrigle HH, erectile dysfunction. J Sex Med context: clinical and basic aspects.
Lees WR, Ralph DJ, Neild GH 7:19181927 In: Dennerstein L, Fraser I (eds)
(2004) A novel therapy with Corona G, Monami M, Boddi V, Hormones and Behavior. Elsevier
testosterone and sildenafil for Cameron-Smith M, Fisher AD, de Science Publishers, New York, pp
erectile dysfunction in patients on Vita G, Melani C, Balzi D, Sforza A, 433441
renal dialysis or after renal Forti G, Mannucci E, Maggi M Dettenhofer MH (2009) Women,
transplantation. J Fam Plann Reprod (2010c) Low testosterone is eunuchs and imperial courts. In:
Health Care 30:8890 associated with an increased risk of Sheidel W (ed) Rome and China:
Corona G, Maggi M (2010) The role of MACE lethality in subjects with Comparative Perspectives on Ancient
testosterone in erectile dysfunction. erectile dysfunction. J Sex Med World Empires. Oxford University
Nat Rev Urol 7:4656 7:15571564 Press, Oxford, pp 8389

263
Chapter 12: Testosterone and erection

Ellis WJ, Grayhack JT (1963) Sexual Garban H, Marquez D, Cai L, Rajfer J, Jain P, Rademaker AW, McVary KT
function in aging males after Gonzalez-Cadavid NF (1995b) (2000) Testosterone
orchidectomy and estrogen therapy. Restoration of normal adult penile supplementation for erectile
J Urol 89:895899 erectile response in aged rats by dysfunction: results of a meta-
Fabbri A, Jannini EA, Ulisse S, long-term treatment with analysis. J Urol 164:371375
Gnessi L, Moretti C, Frajese G, androgens. Biol Reprod Jannini EA, Screponi E, Carosa E,
Isidori A (1988) Low serum 53:13651372 Pepe M, Lo Giudice F, Trimarchi F,
bioactive luteinizing hormone in Golden SH, Robinson KA, Saldanha I, Benvenga S (1999) Lack of sexual
nonorganic male impotence: Anton B, Ladenson PW (2009) activity from erectile dysfunction is
possible relationship with altered Clinical review: prevalence and associated with a reversible
gonadotropin-releasing hormone incidence of endocrine and reduction in serum testosterone. Int
pulsatility. J Clin Endocrinol Metab metabolic disorders in the J Androl 22:385392
67:867875 United States: a comprehensive Kalinchenko SY, Kozlov GI,
review. J Clin Endocrinol Metab Gontcharov NP, Katsiya GV (2003)
Feldman HA, Goldstein I,
94:18531878 Oral testosterone undecanoate
Hatzichristou DG, Krane RJ,
McKinlay JB (1994) Impotence and Greenstein A, Plymate SR, Katz PG reverses erectile dysfunction
its medical and psychosocial (1995) Visually stimulated associated with diabetes mellitus
correlates: results of the erection in castrated men. J Urol in patients failing on sildenafil
Massachusetts Male Aging Study. 153:650652 citrate therapy alone. Aging Male
J Urol 151:5461 Greenstein A, Mabjeesh NJ, Sofer M, 6:9499
Filippi S, Vignozzi L, Morelli A, Kaver I, Matzkin H, Chen J (2005) Karacan I, Hirshkowitz K (1991) Aging
Chavalmane AK, Sarchielli E, Does sildenafil combined with and sleep apnea in impotent men
Fibbi B, Saad F, Sandner P, testosterone gel improve erectile with arterial risk factors. In: Smirne
Ruggiano P, Vannelli GB, Mannucci E, dysfunction in hypogonadal men in S, Franceschi M, Ferini-Strambi L
Maggi M (2009) Testosterone partially whom testosterone supplement (eds) Sleep and Ageing. Masson,
ameliorates metabolic profile and therapy alone failed? J Urol Milan, pp 135148
erectile responsiveness to PDE5 173:530532 Khler TS, Kim J, Feia K, Bodie J,
inhibitors in an animal model of male Heim N, Hursch CJ (1979) Castration Johnson N, Makhlouf A, Monga M
metabolic syndrome. J Sex Med for sex offenders: treatment or (2008) Prevalence of androgen
6:32743288 punishment? A review and critique deficiency in men with erectile
Fisher AD, Corona G, Bandini E, of recent European literature. Arch dysfunction. Urology 71:693697
Mannucci E, Lotti F, Boddi V, Sex Behav 8:281304 Kraemer HC, Becker HB, Brodie HK,
Forti G, Maggi M (2009) Herndon JG, Perachio AA, Turner JJ, Doering CH, Moos RH, Hamburg
Psychobiological correlates of Collins DC (1981) Fluctuations in DA (1976) Orgasmic frequency and
extramarital affairs and differences testosterone levels of male rhesus plasma testosterone levels in normal
between stable and occasional monkeys during copulatory activity. human males. Arch Sex Behav
infidelity among men with Physiol Behav 26:525528 5:125132
sexual dysfunctions. J Sex Med
Hirshkowitz M, Schmidt MH (2005) Kwan M, Greenleaf WJ, Mann J,
6:866875
Sleep-related erections: clinical Crapo L, Davidson JM (1983) The
Foresta C, Caretta N, Rossato M, perspectives and neural nature of androgen action on male
Garolla A, Ferlin A (2004) Role of mechanisms. Sleep Med Rev sexuality: a combined laboratory-
androgens in erectile function. 9:311329 self-report study on hypogonadal
J Urol 171:23582362 men. Clin Endocrinol Metab
Hwang TI, Chen HE, Tsai TF, Lin YC
Fox CA, Ismail AA, Love DN, Kirkham (2006) Combined use of androgen 57:557562
KE, Loraine JA (1972) Studies on and sildenafil for hypogonadal LaFerla JJ, Anderson DL, Schalch DS
the relationship between plasma patients unresponsive to (1978) Psychoendocrine response to
testosterone levels and human sildenafil alone. Int J Impot Res sexual arousal in human males.
sexual activity. J Endocrinol 18:400404 Psychosom Med 40:166172
52:5158 Isidori AM, Giannetta E, Gianfrilli D, Lee PA, Jaffe RB, Midgley AR Jr (1974)
Garban H, Vernet D, Freedman A, Greco EA, Bonifacio V, Aversa A Lack of alteration of serum
Rajfer J, Gonzalez-Cadavid N (2005) Effects of testosterone on gonadotropins in men and
(1995a) Effect of aging on nitric sexual function in men: results of a women following sexual
oxide-mediated penile erection in meta-analysis. Clin Endocrinol (Oxf) intercourse. Am J Obstet Gynecol
rats. Am J Physiol 268:H467H475 63:381394 120:985987

264
Chapter 12: Testosterone and erection

Levy JB, Seay TM, Tindall DJ, NIH Consensus Conference (1993) Raboch J, Starka L (1973) Reported
Husmann DA (1996) The effects of Impotence. NIH Consensus coital activity of men and levels of
androgen administration on phallic Development Panel on Impotence. plasma testosterone. Arch Sex Behav
androgen receptor expression. JAMA 270:8390 2:309315
J Urol 156:775779 OCarroll R, Shapiro C, Bancroft J Reilly CM, Zamorano P, Stopper VS,
Lindau ST, Schumm LP, Laumann EO, (1985) Androgens, behaviour and Mills TM (1997) Androgenic
Levinson W, OMuircheartaigh CA, nocturnal erection in hypogonadal regulation of NO availability in rat
Waite LJ (2007) A study of sexuality men: the effects of varying the penile erection. J Androl 18:110115
and health among older adults in replacement dose. Clin Endocrinol Rhoden EL, Telken C, Sogari PR,
the United States. New Engl J Med (Oxf) 23:527538 Souto CA (2002) The relationship
357:762774 Palese MA, Crone JK, Burnett AL of serum testosterone to erectile
Lugg J, Ng C, Rajfer J, Gonzalez- (2003) A castrated mouse model of function in normal aging men.
Cadavid N (1996) Cavernosal nerve erectile dysfunction. J Androl J Urol 167:17451748
stimulation in the rat reverses 24:699703 Rochira V, Zirilli L, Madeo B,
castration-induced decrease in Park KH, Kim SW, Kim KD, Paick JS Balestrieri A, Granata AR, Carani C
penile NOS activity. Am J Physiol (1999) Effects of androgens on the (2003) Sex steroids and sexual desire
271:E354E361 expression of nitric oxide synthase mechanism. J Endocrinol Invest
Lugg JA, Rajfer J, Gonzalez-Cadavid mRNAs in rat corpus cavernosum. 26:2936
NF (1995) Dihydrotestosterone is BJU Int 83:327333 Rochira V, Balestrieri A, Madeo B,
the active androgen in the Penson DF, Ng C, Cai L, Rajfer J, Granata AR, Carani C (2006)
maintenance of nitric oxide- Gonzalez-Cadavid NF (1996) Sildenafil improves sleep-related
mediated penile erection in the rat. Androgen and pituitary control of erections in hypogonadal men:
Endocrinology 136:14951501 penile nitric oxide synthase and evidence from a randomized,
Marin R, Escrig A, Abreu P, Mas M erectile function in the rat. Biol placebo-controlled, crossover study
(1999) Androgen-dependent nitric Reprod 55:567574 of a synergic role for both
oxide release in rat penis correlates Penson DF, Ng C, Rajfer J, Gonzalez- testosterone and sildenafil on
with levels of constitutive nitric Cadavid NF (1997) Adrenal penile erections. J Androl
oxide synthase isoenzymes. Biol control of erectile function and 27:165175
Reprod 61:10121016 nitric oxide synthase in the rat Roffwarg HP, Sachar EJ, Halpern F,
Mills TM, Wiedmeier VT, Stopper VS penis. Endocrinology 138: Hellman L (1982) Plasma
(1992) Androgen maintenance of 39253932 testosterone and sleep: relationship
erectile function in the rat penis. Petak SM, Nankin HR, Spark RF, to sleep stage variables. Psychosom
Biol Reprod 46:342348 Swerdloff RS, Rodriguez-Rigau LJ; Med 44:7384
Montorsi F, Oettel M (2005) American Association of Clinical Rosen R, Altwein J, Boyle P, Kirby RS,
Testosterone and sleep-related Endocrinologists (2002) American Lukacs B, Meuleman E, OLeary
erections: an overview. J Sex Med Association of Clinical MP, Puppo P, Robertson C,
2:771784 Endocrinologists Medical Giuliano F (2003) Lower urinary
Morelli A, Filippi S, Mancina R, Guidelines for clinical practice for tract symptoms and male sexual
Luconi M, Vignozzi L, Marini M, the evaluation and treatment of dysfunction: the multinational
Orlando C, Vannelli GB, Aversa A, hypogonadism in adult male survey of the aging male (MSAM-7).
Natali A, Forti G, Giorgi M, Jannini patients 2002 update. Endocr Pract Eur Urol 44:637649
EA, Ledda F, Maggi M (2004) 8:440456 Rosen RC, Riley A, Wagner G,
Androgens regulate Petrone L, Mannucci E, Corona G, Osterloh IH, Kirkpatrick J, Mishra
phosphodiesterase type 5 Bartolini M, Forti G, Giommi R, A (1997) The international index of
expression and functional activity in Maggi M (2003) Structured erectile function (IIEF): a
corpora cavernosa. Endocrinology interview on erectile dysfunction multidimensional scale for
145:22532263 (SIEDY): a new, multidimensional assessment of erectile dysfunction.
Morelli A, Corona G, Filippi S, instrument for quantification of Urology 49:822830
Ambrosini S, Forti G, Vignozzi L, pathogenetic issues on erectile Rosenthal BD, May NR, Metro MJ,
Maggi M (2007) Which patients dysfunction. Int J Impot Res Harkaway RC, Ginsberg PC (2006)
with sexual dysfunction are suitable 15:210220 Adjunctive use of AndroGel
for testosterone replacement Podlasek CA (2009) Sonic hedgehog, (testosterone gel) with sildenafil to
therapy? J Endocrinol Invest apoptosis, and the penis. J Sex treat erectile dysfunction in men
30:880888 Med 6 Suppl 3:334339 with acquired androgen deficiency

265
Chapter 12: Testosterone and erection

syndrome after failure using sildenafil in male dialysis patients sexual activity. J Endocrinol Invest
sildenafil alone. Urology 67:571574 with erectile dysfunction 28:3944
Schirar A, Chang C, Rousseau JP unresponsive to erythropoietin and/ Vignozzi L, Morelli A, Filippi S,
(1997) Localization of androgen or testosterone treatments. Int Ambrosini S, Mancina R, Luconi M,
receptor in nitric oxide synthase- J Impot Res 18:6168 Mungai S, Vannelli GB, Zhang XH,
and vasoactive intestinal peptide- Traish AM (2009) Androgens play a Forti G, Maggi M (2007)
containing neurons of the major pivotal role in maintaining penile Testosterone regulates RhoA/Rho-
pelvic ganglion innervating the rat tissue architecture and erection: kinase signaling in two distinct
penis. J Neuroendocrinol 9:141150 a review. J Androl 30:363369 animal models of chemical diabetes.
Traish AM, Park K, Dhir V, Kim NN, J Sex Med 4:620630
Shabsigh R, Raymond JF, Olsson CA,
OToole K, Buttyan R (1998) Moreland RB, Goldstein I (1999) Vignozzi L, Filippi S, Morelli A,
Androgen induction of DNA Effects of castration and androgen Marini M, Chavalmane A, Fibbi B,
synthesis in the rat penis. Urology replacement on erectile function in Silvestrini E, Mancina R, Carini M,
52:723728 a rabbit model. Endocrinology Vannelli GB, Forti G, Maggi M
140:18611868 (2009) Cavernous neurotomy in the
Shabsigh R, Kaufman JM, Steidle C,
Traish AM, Munarriz R, OConnell L, rat is associated with the onset of an
Padma-Nathan H (2004)
Choi S, Kim SW, Kim NN, Huang overt condition of hypogonadism.
Randomized study of testosterone
YH, Goldstein I (2003) Effects of J Sex Med 6:12701283
gel as adjunctive therapy to
sildenafil in hypogonadal men with medical or surgical castration on Wang C, Nieschlag E, Swerdloff R,
erectile dysfunction who do not erectile function in an animal Behre HM, Hellstrom WJ,
respond to sildenafil alone. J Urol model. J Androl 24:381387 Gooren LJ, Kaufman JM, Legros JJ,
172:658663 Traish AM, Toselli P, Jeong SJ, Kim NN Lunenfeld B, Morales A, Morley JE,
(2005) Adipocyte accumulation in Schulman C, Thompson IM,
Shamloul R, Ghanem H, Fahmy I, Weidner W, Wu FC; International
El-Meleigy A, Ashoor S, Elnashaar A, penile corpus cavernosum of the
orchiectomized rabbit: a potential Society of Andrology; International
Kamel I (2005) Testosterone Society for the Study of Aging Male;
therapy can enhance erectile mechanism for veno-occlusive
dysfunction in androgen deficiency. European Association of Urology;
function response to sildenafil in European Academy of Andrology;
patients with PADAM: a pilot study. J Androl 26:242248
American Society of Andrology
J Sex Med 2:559564 Tsertsvadze A, Fink HA, Yazdi F, (2009) Investigation, treatment, and
Shen ZJ, Zhou XL, Lu YL, Chen ZD MacDonald R, Bella AJ, Ansari MT, monitoring of late-onset
(2003) Effect of androgen Garritty C, Soares-Weiser K, hypogonadism in males: ISA,
deprivation on penile ultrastructure. Daniel R, Sampson M, Fox S, ISSAM, EAU, EAA, and ASA
Asian J Androl 5:3336 Moher D, Wilt TJ (2009) Oral recommendations. Eur Urol
phosphodiesterase-5 inhibitors and 55:121130
Stearns EL, Winter JS, Faiman C (1973) hormonal treatments for erectile
Effects of coitus on gonadotropin, dysfunction: a systematic review Wingard CJ, Johnson JA, Holmes A,
prolactin and sex steroid levels in and meta-analysis. Ann Intern Med Prikosh A (2003) Improved erectile
man. J Clin Endocrinol Metab 151:650661 function after Rho-kinase inhibition
37:687691 in a rat castrate model of erectile
van Anders SM, Hamilton LD, dysfunction. Am J Physiol Regul
Stoleru SG, Ennaji A, Cournot A, Spira Watson NV (2007) Multiple partners Integr Comp Physiol 284:
A (1993) LH pulsatile secretion and are associated with higher testosterone R1572R1579
testosterone blood levels are in North American men and women.
influenced by sexual arousal in Horm Behav 51:454459 Wu FC, Tajar A, Pye SR, Silman AJ,
human males. Finn JD, ONeill TW, Bartfai G,
Vernet D, Cai L, Garban H, Babbitt ML, Casanueva F, Forti G, Giwercman A,
Psychoneuroendocrinology
Murray FT, Rajfer J, Gonzalez-
18:205218 Huhtaniemi IT, Kula K, Punab M,
Cadavid NF (1995) Reduction of Boonen S, Vanderschueren D;
Takahashi Y, Hirata Y, Yokoyama S, penile nitric oxide synthase in European Male Aging Study
Ishii N, Nunes L, Lue TF, Tanagho EA diabetic BB/WORdp (type I) and Group (2008) Hypothalamic-
(1991) Loss of penile erectile response BBZ/WORdp (type II) rats with pituitary-testicular axis disruptions
to intracavernous injection of erectile dysfunction. Endocrinology in older men are differentially linked
acetylcholine in castrated dog. Tohoku 136:57095717 to age and modifiable risk factors:
J Exp Med 163:8591 Vignozzi L, Corona G, Petrone L, the European Male Aging Study.
Tas A, Ersoy A, Ersoy C, Gullulu M, Filippi S, Morelli AM, Forti G, J Clin Endocrinol and Metab
Yurtkuran M (2006) Efficacy of Maggi M (2005) Testosterone and 93:27372745

266
Chapter 12: Testosterone and erection

Wu FC, Tajar A, Beynon JM, Pye SR, in hypogonadal men unresponsive Zhang XH, Morelli A, Luconi M,
Silman AJ, Finn JD, ONeill TW, to tadalafil: results from an open- Vignozzi L, Filippi S, Marini M,
Bartfai G, Casanueva FF, Forti G, label uncontrolled study. Andrologia Vannelli GB, Mancina R, Forti G,
Giwercman A, Han TS, Kula K, 38:6168 Maggi M (2005) Testosterone
Lean ME, Pendleton N, Punab M, Zhang MG, Shen ZJ, Zhang CM, regulates PDE5 expression and in
Boonen S, Vanderschueren D, Wu W, Gao PJ, Chen SW, Zhou WL vivo responsiveness to tadalafil in
Labrie F, Huhtaniemi IT; EMAS (2011) Vasoactive intestinal rat corpus cavernosum. Eur Urol
Group (2010) Identification of late- polypeptide, an erectile 47:409416
onset hypogonadism in middle- neurotransmitter, improves erectile Zhang XH, Filippi S, Morelli A,
aged and elderly men. N Engl J Med function more significantly in Vignozzi L, Luconi M, Donati S,
363:123135 castrated rats than in normal rats. Forti G, Maggi M (2006)
Yassin AA, Saad F (2008) Testosterone BJU 108:440446 Testosterone restores diabetes-
and erectile dysfunction. J Androl Zhang XH, Hu LQ, Zheng XM, Li SW induced erectile dysfunction and
29:593604 (1999) Apoptosis in rat erectile sildenafil responsiveness in two
Yassin AA, Saad F, Diede HE (2006) tissue induced by castration. Asian distinct animal models of chemical
Testosterone and erectile function J Androl 1:181185 diabetes. J Sex Med 3:253264

267
Chapter
Testosterone and the prostate

13 John T. Isaacs and Samuel R. Denmeade

13.1 Introduction 268 13.9 Role of androgen in prostate


13.2 Evolutionary selection of testosterone as carcinogenesis 282
master regulator of male reproduction 270 13.10 Androgen ablation as therapy for metastatic
13.3 Role of testosterone in the development and prostate cancer 284
maintenance of the human prostate 272 13.11 SRD5A inhibitors for the prevention of
13.4 Testosterone metabolism in the prostate 273 prostate cancer 285
13.5 Androgen receptor as integrator of androgen 13.12 New direction for treatment of castration-
signaling in the prostate 275 resistant prostate cancer: bipolar androgen
13.6 Mechanism of androgen action in the therapy 286
prostate 277 13.13 Side-effects of androgen replacement/
13.7 Organization of prostate epithelial and ablation in the aging male 286
stromal stem cell units 279 13.14 Key messages 287
13.8 Androgen in benign prostatic hyperplasia and 13.15 References 287
the clinical use of SRD5A inhibitors for its
treatment 281

13.1 Introduction prostatae. However, he and his contemporaries mis-


During evolution, mammals developed accessory sex interpreted the history of the organ and the term, chose
glands, which in males of the species are named either the wrong gender when translating it into Latin, and
for their anatomical position in adult animals or for their believed that it designated a double organ. Only in the
assumed functions (Price and Williams-Ashman 1961). 1800s was this anatomical error corrected, while the
The only gland present in all orders of male mammals, grammatical one lived on in the term prostate.
even the egg-laying monotremes, is the prostate. The Thus, the gland that in male mammals stands
term prostate has an interesting but confusing history before the base of the bladder and produces and
which has been extensively reviewed by Marx and Kar- releases secretion into the male ejaculate is defined
enberg (2009). According to these authors, in ancient as the prostate. In most male mammals, there are
Greek the masculine term prostats literally meant additional glands that likewise release excretion into
someone who stands before someone or something the ejaculate, and these glands are given a variety of
and is the origin of the term president or principal. names depending on the species. In humans, the male
This term, however, was never used in ancient Greece in a accessory sex tissues consist of the prostate including
medical sense. It was not until the Renaissance that the periurethral glands (aka glands of Littre), seminal
anatomists discovered the prostate, initially naming it vesicles, and bulbourethral glands (Cowpers glands)
the glandulous body. In 1600 the French physician du (Fig. 13.1). Although it is believed that the prostate is
Laurens introduced the metaphoric denomination important in protecting the lower urinary tract from

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

268
Chapter 13: Testosterone and the prostate

Vertebral column
Urinary
bladder Ureter

Pubic Rectum
bone
Seminal vesicle
Ductus
deferens Ejaculatory duct
Prostate gland
Penis

Anus

Cords of
erectile
Bulbourethral gland
tissue
Urethra
Glans
penis
Epididymis
Scrotum
Testis
Fig. 13.1 Cross-section of the adult human male.

infection and for fertility, it is frequently the site of prostatic diseases, the etiologies of neither prostatic
infection and inflammation, and sperm harvested cancer nor BPH are known. A major reason why both
from the epididymis without exposure to seminal or the specific function of the prostate and etiology of the
prostatic fluid can produce fertilization and successful prostatic neoplasms have been difficult to elucidate is
birth (Silver et al. 1988). Thus, the prostate makes that the gross structure and histological appearance of
nothing that is absolutely required for male fertility. this gland vary widely in the animal kingdom, and thus
The fact that the specific function of the prostate is comparative animal studies have been problematic.
not fully understood might not be so problematic if it In fact, no organ system varies so widely among the
were not the case that the prostate is the most common animal species as the male sex accessory tissues (Price and
site of neoplastic transformation in men, with nearly Williams-Ashman 1961) (Fig. 13.2). In contrast to
one million men dying of prostate cancer globally each humans, rats and mice have a prostate that is composed
year (Jemal et al. 2011). Furthermore, the prostate is of four anatomically and biochemically distinct prostatic
the most common site of benign neoplastic disease in lobes (i.e. the ventral, dorsal, lateral and anterior lobes: the
males (Berry et al. 1984). More than 50% of all men last of these also called the coagulating gland). In addition,
above the age of 50 have benign prostatic hyperplasia these species have seminal vesicles and preputial glands.
(BPH), with 25% of men eventually requiring treat- Many species (e.g. primates (monkeys) and marsupials
ment for this condition (Berry et al. 1984). Thus, it is like opossum) have a segmented prostate and seminal
remarkable that, despite the high prevalence of vesicles (Fig. 13.2). Bulls have very small prostates and

269
Chapter 13: Testosterone and the prostate

Fig. 13.2 Comparative anatomy of male accessory sex tissue in a series of mammals (rat, opossum, bull, dog, monkey and man).
Abbreviations: BL or B, bladder; U or UR, urethra; P or PR or PG, prostate; S or SV, seminal vesicle; BU or BG, bulbourethral gland; D or AG,
ampulla of ductus (vas) differens; T, testis; CG, coagulating gland (aka anterior prostate lobe); VP, ventral prostate lobe; LP, later prostate lobe;
DP, dorsal prostate lobe; VD, vas deferens.

large seminal vesicles (Fig. 13.2). The normal human prostatic cells has allowed the abnormal detection of these
prostate is not composed of anatomically separate or proteins in the serum of men to be useful as a means of (1)
segmented lobes as in many animals, but is instead initially detecting prostatic cancer in asymptomatic men,
divided into four zones (Fig. 13.3). The peripheral zone (2) monitoring residual presence of systemic micrometa-
comprises 70 to 75% of the gland, the central zone 20 to static disease in men who have undergone radical prosta-
25%, and the transitional zone 5%, while the anterior tectomy for presumed localized disease, and (3)
surface consists of the fibromuscular stroma (McNeal monitoring the response of clinically detected metastatic
et al. 1988). Most cancers develop in the peripheral zone. disease to systemic therapy. Although other animal
Benign prostatic hyperplasia develops in the transitional species synthesize prostate-specific proteins (e.g. prosta-
zone as a part of the aging process. In contrast, the dog has tein and probasin in the rat and the arginine esterase in
a well-developed prostate which has no zonal anatomy the dog), there are no genes directly homologous to PSA
and completely lacks seminal vesicles (Fig. 13.2). Import- or HK2, based on DNA sequence, in the dog or rat
antly, the dog is the only species other than man which genome (Olsson et al. 2004). There is a homologous
spontaneously develops both BPH and prostatic cancer prostatic-specific acid phosphatase gene in the rat; how-
with aging (Isaacs 1984). Besides this anatomical vari- ever, the level of expression is nearly 1000-fold lower in rat
ation, there is a large variation among the different species versus human prostate epithelial cells (Coffey 1992).
in the secretory products produced and released by the
prostate into the ejaculate (Mann and Mann 1981).
For example, the human prostatic epithelial cells syn- 13.2 Evolutionary selection of
thesize and secrete a series of unique proteins into the testosterone as master regulator of
ejaculate (Coffey 1992). These include serine protease,
PSA, human glandular kallikrein-2 (HK2) and prostatic- male reproduction
specific acid phosphatase. The essentially exclusive pro- Based on such varied anatomy and biochemistry, it
duction of these proteins by normal and malignant has been difficult to utilize animal models to define

270
Chapter 13: Testosterone and the prostate

Fig. 13.3 Zonal anatomy of the human


prostate.

Central zone

Anterior
fibromuscular
stroma

Ejaculatory Transition zone


duct

Peripheral zone

Urethra

the etiology of either BPH or prostatic cancer. It Thornton 2011). In fact, the presence of testoster-
is known, however, that during mammalian evolu- one may have been an important driving factor
tion, testosterone was selected as the master regula- for the development of the AR, which is the last
tor for the embryonic development and postnatal of the steroid receptors to have evolved (Eick and
growth and maintenance of the prostate and the Thornton 2011).
other male accessory sex tissues, as well as sperm- Another driving factor involved in the selective
atogenesis, bone and muscle maturation and libido. pressure for testosterone becoming the master regu-
Why testosterone was selected during mammalian lator of reproduction during evolution relates to the
evolution as the master regulator for male physi- unique danger of reproduction in mammals. During
ology and reproduction is not fully known. It is mammalian evolution, breeding became episodically
known that during evolution, all the members of timed for optimal survival of both parents and
the steroid receptors (i.e. estrogen (ERa and ERb), offspring. Thus, as well as reduced sexual libido and
mineralocorticoid (MR), glucocorticoid (GR), pro- sperm maturation, the male accessory sex glands are
gesterone (PR), and androgen (AR)) descended atrophic during the seasonal period when breeding
from a single ancestral receptor which branched is not optimal. When environmental conditions (i.e.
off from the rest of the nuclear receptor (NR) food/temperature/light etc.) are appropriate, male
superfamily early in animal evolution (Eick and mammals come into breeding season. Such seasonal
Thornton 2011). The similarity of all the inverte- breeding has great reproductive advantage and was
brate receptor sequences to the vertebrate ERs sug- selected during evolution to restrict the energy
gests that the ancestral steroid receptor protein was requirements for maintaining male accessory sex
likely to have been ER-like in both sequence and in glands, sperm maturation, and sexual libido, thus
function, and that the other steroid receptors pro- limiting the dangers associated with the maniacal
liferated and diversified through a series of its gene focus upon procreation at the expense of self-survival
duplications. Phylogenetic studies have documented (i.e. suppression of normal flight instinct in the pres-
that the enzymes of estrogen synthesis, which con- ence of predators) to the minimal time frame of
verts cholesterol to progesterone to testosterone to the breeding season. Since the breeding season
estrogen, co-evolved with the ER receptors and were for many mammals is short, evolutionary pressure
present before the evolution of the steroid receptors drove development of a neuroendocrine (i.e. pineal
sensitive to these latter two steroids (Eick and gland-hypothalamic-pituitary) axis to restrict high

271
Chapter 13: Testosterone and the prostate

Fig. 13.4 Serum testosterone levels


during various stages of development
and aging in human males.
A Annual B Daily C Pulsatile
800 800 800
600 600 600
400 400 400
200 200 200
3 6 9 6 12 18 2 4 6 8 10
Months Hours Hours

600
Testosterone (ng / dl)

A
400 C
B

200

100

0 Fetal Neo Pre Pubertal Adult Senescence


natal pubertal

production and secretion of testosterone into the cir- 13.3 Role of testosterone in the
culation by the testes to only the breeding season.
Coincident with the evolution of seasonal breed- development and maintenance of the
ing, there was also selective pressure to develop a human prostate
mechanism for the rise of testosterone to rapidly The urogenital sinus derived from the endoderm
induce sexual libido, sperm maturation and full layer is the embryonic anlagen from which the
growth of the atrophic male accessory sex tissues prostate develops in utero. For the human prostate
needed for reproduction during the short breeding to develop normally, a critical level of androgenic
season. Thus during evolutionary selection of testos- stimulation is required at specific times during its
terone as the master regulator of seasonal male development in utero (Wilson 1984). In the develop-
reproduction, there was co-selection of enzymes ing male, the fetal testis secretes testosterone into
needed to regulate intracellular ligand and the AR the fetal circulation at levels sufficient to stimulate
needed to generate intracellular signaling induced the differentiation and growth of a portion of the
by its ligand binding (Eick and Thornton 2011). In urogenital sinus tissue, producing the definitive pro-
contrast with the other mammals, human males state gland. In humans, this begins during the first
eventually evolved away from strict seasonal breed- three months of fetal growth and remains high until
ing and acquired the ability to sustain serum testos- the last months of pregnancy (Fig. 13.4). If sufficient
terone at a sufficiently high chronic level to maintain serum testosterone is not present at this critical state
the male accessory sex glands, spermatogenesis and of intrauterine development, the prostate does not
libido in a fully developed adult state so that fertility develop (Wilson 1984).
is constant. Such constant fertility is a definitive During the first several months after birth, serum
advantage for the highly mobile human species. This testosterone again rises before decreasing to a low
is because it enables reproduction to occur despite baseline value (Fig. 13.4). This neonatal spike in serum
environment restrictions, allowing man to populate testosterone is critical for male imprinting of the
all of the biological niches throughout the world as hypothalamic-pituitary-testicular axis begun during
opposed to locking the species to specific indigenous embryonic development (Gorski 2002; Grumbach
breeding ranges, like other mammals. Apparently, the 2002). After this neonatal period, serum testosterone
price of such reproductive freedom, however, is remains low in humans until puberty when levels rise
the acquisition of BPH and prostate cancer by the to the adult range (Frasier et al. 1969) (Fig. 13.4). Until
human male.

272
Chapter 13: Testosterone and the prostate

puberty, the human prostate remains small (approxi- mechanisms responsible for how testosterone stimu-
mately <2 g; Isaacs 1984). During puberty, the lates such developmental and growth effects, an
human prostate grows to its adult size of approxi- understanding of its metabolism and the role of the
mately 20 g (lsaacs 1984). Between the age of 10 and AR as an integrator of intracellular androgen signal-
20 years, the rate of prostatic growth is exponential, ing is required.
with a prostatic weight-doubling time of 3 years
(lsaacs 1984). This period of exponential growth cor- 13.4 Testosterone metabolism in
responds to the time period when the serum testoster-
one level is rising from initially low levels seen before the prostate
the age of 10 to the high levels seen in an adult human The hypothalamic-pituitary-testicular axis regulates the
male (Frasier et al. 1969; Fig. 13.4). If a boy is cas- serum level of testosterone via a series of feedback
trated before the age of 10, the serum testosterone loops resulting in the testes secreting into the blood
levels do not rise to their normal adult level, and the 6 mg of testosterone per day (Coffey 1992). In the
proliferative growth of the human prostate between serum, the total testosterone is 20 nM, with 98% of
10 and 20 years of life is completely blocked (Moore the testosterone bound to a series of high-affinity (i.e.
1944; Huggins and Johnson 1947). These results dem- testosterone binding protein (TeBG)) or low-affinity
onstrate that a physiological level of testosterone is (i.e. albumin) binding proteins. The 2% non-protein
chronically required for the normal growth of the bound (i.e. free testosterone) level is 0.4 nM, and it
human prostate. This chronic requirement for testos- is this free testosterone which is able to diffuse from
terone derives from the necessity for androgens to the blood into the extracellular fluid of the prostatic
regulate the total prostatic cell number by affecting stromal compartment into the prostate epithelial cells
both the rate of cell proliferation and of cell death. (Coffey 1992). Within these epithelial cells, testosterone
Androgen does this by stimulating the rate of cell is enzymatically converted to 5a-dihydrotestosterone
proliferation (i.e. agonistic ability of androgen) while (Bruchovsky and Wilson 1968). The enzymes respon-
simultaneously inhibiting the rate of cell death (antag- sible for the irreversible conversion of testosterone to
onistic ability of androgen) (Kyprianou and Isaacs DHT are the membrane-bound NADPH-dependent
1988). Because of this dual agonist/antagonist effect D4-3-ketosteroid 5a-oxidoreductases (i.e. SRD5A, aka
of androgen on the prostate, the rate of cell prolifer- 5a-reductases) (Fig. 13.5; Bruchovsky and Wilson
ation is greater than the rate of cell death during the 1968). Phylogenetic analysis of proteins with a steroid
normal prostatic growth period occurring between 10 5a-reductase domain from multiple species indicates
and 20 years of age. Having reached its maximum that this SRD5A family can be separated in to three
adult size by 20 years of age, the prostate normally main groups consisting of: (1) the SRD5A1-SRD5A2
ceases its continuous net growth (lsaacs 1984). This group; (2) the SRD5A3 group containing the human
does not mean, however, that the cells of the adult ortholog to the yeast DFG10 gene; and (3) the GPSN2-
prostate in men over 20 years of age do not continu- SRD5A2L2 group, supporting the idea that different
ously turn over with time, but that the rate of pro- classes of lipids can be substrates for these enzymes and
static cell proliferation is balanced by an equal rate of suggesting that the substrate of the enzyme encoded by
prostatic cell death, such that neither involution nor the common ancestral gene was potentially not a ster-
overgrowth of the gland normally occurs with time. oid (Cantagrel et al. 2010).
Thus the adult prostate in men over 20 is an example Within the prostate, SRD5A13 isoforms are
of a steady-state self-renewing tissue. If an adult male expressed. Each of the isozymes has been cloned and
whose prostate is in this steady-state maintenance the complete DNA-based sequence and amino acid
condition is castrated, serum testosterone levels rap- composition are known (Andersson and Russell 1990;
idly decrease to low values comparable to those seen Jenkins et al.1991; Labrie et al. 1992; Thigpen et al.
in males younger than 10 years of age. As a result, the 1992; Uemura et al. 2008). The genes encoding the
prostate rapidly involutes (Peters and Walsh 1987). proteins for all of the SRD5A isozymes have a similar
Such involution demonstrates that a physiological structure containing five exons separated by four
level of testosterone is chronically required, not introns. The three genes share approximately 46%
only for initial development, but also for maintenance sequence homology and encode for a protein of 29
of the normal prostate. To define the molecular 000 kDa molecular weight. The SRD5A1 isozyme is

273
Chapter 13: Testosterone and the prostate

CYP17A1 CYP17A1 Fig. 13.5 Overview of the metabolic


Cholesterol Pregnenolone 17-Hydroxypregnenolone Dehydroepiandrosterone pathway for conversion of cholesterol to
testosterone, indicating the role of
CYP17A1 CYP17A1 CYP17A1 in this conversion and the
Progesterone 17-Hydroxyprogesterone Androstenedione 5a-reductase-catalyzed conversion
to DHT.

OH OH

5-reductase,
type 1 or type2

O O
H

Dihydrotestosterone Testosterone

encoded by a gene on human chromosome 5p15 secretory/luminal epithelial cells or prostate stromal
(Jenkins et al. 1991). It has a neutral pH optimum, a cells (Godoy et al. 2011). While SRD5A3 can reduce
requirement for high concentration of testosterone to testosterone to DHT, its major normal function
saturate the enzyme (high Km = 3 mM), and is rather appears to be in anabolic metabolism, where it
insensitive to finasteride inhibition (Ki 300 nM) reduces the isoprene unit of polyprenols to form
(Andersson and Russell 1990; Jenkins et al. 1991). dolichols, required for synthesis of dolichol-linked
The type 1 isozyme is expressed at low levels by the monosaccharides, and the oligosaccharide precursor
prostate basal epithelial cells and is the predominant used for N-glycosylation (Cantagrel et al. 2010).
5a-reductase isozyme in skin; it is also present in the Once formed via the 5-reductase, DHT can revers-
liver (Jenkins et al. 1992; Normington and Russell ibly bind to the AR, which will be discussed in the
1992; Berman and Russell 1993). next section, to regulate prostatic cellular prolifer-
The SRD5A2 isozyme is encoded by a gene on ation and survival. Alternatively, DHT can be further
human chromosome 2p23 (Thigpen et al. 1992). It reductively metabolized to 5x-androstane-3,17-diol
has an acidic (pH 5.0) optimum and has a lower (3a-diol) by the 3a-hydroxysteroid dehydrogenase
Km (0.5 mM) for testosterone. The SRD5A2 isozyme type 3 isozyme (i.e. 3a-HSD type 3 enzyme, aka
is the predominant 5a-reductase in androgen target AKR1C2) (Rizner et al. 2003; Fig. 13.6). Once formed,
tissue, including the prostate, where it is expressed in 3a-diol can he re-oxidized back to DHT via an oxida-
the stromal cells (Berman and Russell 1993; Bayne tive 3a-HSD enzyme not fully characterized in the
et al. 1998). Analysis of individuals with male normal prostate, or glucuronidated at position 3 and
pseudo-hermaphroditism caused by 5a-reductase excreted by the prostate (Rizner et al. 2003). 3a-Diol
deficiency has revealed no mutation in the type 1 can also be oxidized at its l7b-hydroxy position
isozyme gene (Jenkins et al. 1992). In contrast, by 17b-HSD type 2 or type 6 isozymes to form
molecular analysis demonstrated that mutation in 5a-androsterone, or can also be glucuronidated at
the SRD5A2 gene accounts for this disorder (Thigpen position 3 and excreted (Biswas and Russell 1997;
et al. 1992). Based on these results, it has been sug- Rizner et al. 2003; Fig. 13.6). Dihydrotestosterone can
gested that the SRD5A1 isozyme functions in a cata- also be either oxidatively metabolized at its l7b-hydroxy
bolic manner in the metabolic removal of androgens group by the 17b-HSD type 2 isozyme to form
by non-target tissue, whereas SRD5A2 isozyme func- 5a-androstane-3,17-dione (Rizner et al. 2003), or
tions in an anabolic role to amplify the androgenicity reductively metabolized at its 3-keto group to produce
of testosterone by effectively converting it to DHT 5a-androstane-3,17b-diol (3b-diol) by l7b-HSD type 7
within androgen target tissue (Normington and Rus- isozyme (Torn et al. 2003; Fig. 13.6). Interestingly, it
sell 1992). The SRD5A3 isoform is encoded by a gene has been documented that the endogenous estrogen
on human chromosome 4q12 (Cantagrel et al. 2010) in the prostate is not l7b-estradiol but 3b-diol
and is normally expressed in prostate basal, but not (Weihua et al. 2001). Also it has been documented

274
Chapter 13: Testosterone and the prostate

Glucuronidation

17-HSD
3-diol Androsterone
Type 2 and 6
3-HSD 3-HSD
Type 3 Type ? 17-HSD Androstane-3,17-dione

Type 2
5-reductase
Testosterone DHT 17-HSD CYP7B1 6-and 7-
Type 1 and 2 3-diol
triol
Type 7

Estrogen
Androgen
receptor-
receptor

Proliferation

Fig. 13.6 Summary of enzymatic pathway of androgen within the human prostate.

that, within the normal prostate, both the isoforms complete androgen insensitivity syndrome and no
of the estrogen receptor (i.e. ERa and ERb) are prostate develops (Gottlieb et al. 1999). AR is a
expressed and both bind 3b-diol (Weihua et al. 2001). ligand-dependent zinc finger DNA-binding protein
The ERa is expressed predominately in the prostatic whose genomic binding coordinates formation of
stromal cells, while ERb is expressed in the epithelial transcriptional complexes at the regulatory elements
cells (Fixemer et al. 2003). The Gustafsson group of targeted genes. The AR gene is located on the long
which initially discovered ERb has postulated that arm of the X chromosome (i.e. Xq11.2) and encodes a
3b-diol binding to the ERb within prostatic epithelial protein with three critical domains: (1) an N-terminal
cells results in antagonism of the AR signaling for domain involved in homotypic dimerization and
epithelial cell proliferation (Weihua et al. 2002). binding with other transcriptional coactivator or co-
The level of such an ER-dependent anti-proliferative repressor proteins; (2) a DNA-binding domain with
effect is thus dependent upon the level of 3b-diol. two zinc finger binding motifs and hinge region; and
This 3b-diol level is itself regulated by the activity (3) a C-terminal steroid LBD, which is also involved
of the CYP7B1 enzyme which hydroxylates 3b-diol in homotypic dimerization and coactivation binding
to 5a-androstane-3b,6a,17b-triol (6a-triol) and 5a- (Dehm and Tindall 2007; Fig. 13.7). This C-terminal
androstane-3b,7a,17b-triol (7a-triol) (lsaacs et al. LBD is also where 90 kDa heat shock protein (i.e.
1979; Weihua et al. 2002; Fig 13.6). HSP90) dimers bind to stabilize the AR protein
during its folding, subsequent to its synthesis. Specific
13.5 Androgen receptor as integrator interaction with androgenic ligands results in the
conformational activation of AR. This allows the dis-
of androgen signaling in the prostate sociation of the HSP90 dimer proteins and thus the
Androgen receptor is the integrator of androgen sig- binding and dimerization of the occupied AR to AREs
naling in the prostate, as documented by the fact that present in the promoter and enhancer regions in
germline truncation mutations early in the first exon AR-regulated genes (Fig. 13.8; Wang et al. 2005). This
of the gene preventing expression of AR result in initial genomic AR binding allows further binding

275
Chapter 13: Testosterone and the prostate

Xp Xq Fig. 13.7 Organization of the AR gene,


mRNA and protein. AR is coded by a
180 kb gene located on the long arm of
q11.2 q12 the X chromosome (i.e. 11q11.2) and,
hence, is present as a single gene copy
5 1 2 3 4 5 6 7 8 3 AR gene per cell. Upper panel indicates the
position of the 8 exons of AR gene mRNA;
180 kb
middle panel indicates which exon
encodes which base pairs (bp) in mRNA;
3564 AAAA and the lower panel aligns the individual
2888 3289 3876 bp exons with the encode portion of the AR
ATG
protein.
m7Gppp 1116 bp- 3436
2731 3000 3722 TGA
AR mRNA
translation
start site 4.3 kb

N-terminal transactivating domain DBD HR Ligand-binding domain

1 559 625 672 919 AR protein


NLS2
(Gln)n (Pro)n (Gly)n NLS1 amino 110 kDa
HSP90 acids

Cooperative binding and


DNA-dependent dimerization

AR ARE DNA AR

Enhancer

AR AR

SRC-1/p160

CBP/P300

SRC1-/p160 Polymerase II
PCAF
TFIIF
Transcription
TFIIF
AR AR

Histones
Promoter
HAT activity

Fig. 13.8 Overview of transcriptional complex organized by AR at the ARE of the promoter/enhancer region of the PSA gene. The AR
assembles a transcription initiation complex on the promoter/enhancer regions of androgen-regulated genes like PSA. Ligand-bound AR
undergoes a DNA-dependent dimerization in the nucleus and recruits a number of transcription factors that acetylate histones, recruit
additional transcription factors and DNA polymerase, and initiate transcription of the target gene.

276
Chapter 13: Testosterone and the prostate

to specific regions of the bound AR by additional and regulate the function of the AR (Liao et al.
nuclear proteins (i.e. transcriptional coactivator pro- 1972; Dehm and Tindall 2007).
teins like SRC-1, ARA 70, etc., and general trans-
cription factors like TFII H and F) to produce 13.6 Mechanism of androgen action
transcriptional complexes that can activate or repress
specific gene expression. For activation, formation in the prostate
of an active transcriptional complex is required, To understand the mechanism for AR-dependent
resulting in site-directed chromatin remodeling via signaling in prostate, an understanding of the two
histone acetylation and methylation that enhances phases of the development of the prostate is needed.
target gene expression (Fig. 13.8). SRC-1 is a member The first ontogeny phase is initiated during embry-
of the p160 transcriptional coactivator gene family that onic life and continues into the early neonate period.
includes SRC-1, TIF2 (also termed GRIP-1 and SRC-2), During embryonic life, the prostate develops from the
and p/CIP (also termed RAC3, ACTR, AIBI and endodermal derived UGS anlagen in males under
SRC-3). Cell-free in-vitro transcription and in-vivo the stimulation of systemic androgen from the testes
experiments have indicated that the SRC-1 family (Fig. 13.3). Such ontogeny requires reciprocal para-
members enhance AR-dependent transactivation of crine interactions between UGS stromal cells and
nuclear genes; the mechanism for such enhancement UGS epithelial cells, driven by a critical level of circu-
involves binding of p160 proteins to the DNA-bound lating androgen (Fig. 13.9). Prostate ontogeny is ini-
AR (Fig. 13.8). This allows the p160 to acetylate his- tiated by an epithelial-to-stroma paracrine interaction
tones via its histone acetyltransferase (HAT) activity. permanently committing UGS mesenchymal stem
Additional coactivators with histone acetyltransferase cells denoted no. 1 (Fig. 13.9), to producing a subset
activity such as CBP, p300 or p/CAF also bind to the of differentiated progeny denoted no. 2 (Fig. 13.9),
p160/AR complex. This results in chromatin remodel- which mature into smooth muscle cells (SMC)
ing and additional binding of general transcription denoted no. 3 (Fig. 13.9), expressing both SRD5A2
factors such as TFIIH and TFIIF with the AR coactiva- enzyme and AR protein (Isaacs 2008).
tion complexes (Litvinov et al. 2003; Dehm and Tindall The commitment of UGS stromal stem cells
2007; Fig. 13.8). to produce SMC expressing both of these proteins is
Both testosterone and DHT can bind to AR; how- dependent upon paracrine factors (i.e. Sonic hedge-
ever, within the prostate, the metabolic pathways hog, etc.) secreted by ectoderm UGS epithelial cells
described in Fig. 13.6 are regulated so that DHT is denoted no. 4 (Fig. 13.9) (Isaacs 2008). This induction
present at a fivefold molar excess compared to testos- is mandatory since genetically inherited defects in
terone (i.e. 10 nM for DHT vs. 2.0 nM for testosterone; either SRD5A2 or AR prevent prostate development
Page et al. 2011). In addition, DHT has nearly a 10-fold (Litvinov et al. 2003). This is because expression of
higher AR binding affinity than testosterone (i.e. Kd SRD5A2 by these SMC allows them to irreversibly
of 0.1 nM for DHT vs. 1.0 nM for testosterone) convert testosterone to DHT. Dihydrotestosterone
(Litvinov et al. 2003). Thus, with regard to prostate is 10 times more potent than testosterone in its ability
physiology, testosterone is actually a prohormone, to stimulate AR-dependent transcription (Litvinov
with DHT being the active intracellular androgenic et al. 2003). Thus, this 5a-reductase activity amplifies
hormone. The extensive metabolic pathway for andro- the low levels of circulating androgen secreted by
gen within the prostate (Fig. 13.6) functions as a means the embryonic testes to produce a sufficient level
for autoregulation, so that the prostatic level of DHT of DHT to bind and activate AR signaling in these
remains constant despite fluctuations in serum testos- SMC, denoted no. 5 (Fig. 13.9). This AR signaling
terone. This is significant since there are episodic and stimulates synthesis and secretion of soluble para-
diurnal variations in both total and free serum testos- crine and autocrine growth factors (GF) and survival
terone levels in humans (Plymate et al. 1989; Fig. 13.3). factors (SF), termed andromedins (e.g. IGF-1, FGF-7
Because growth versus regression (i.e. death) of the and-10, and VEGF) by these SMC, denoted no. 6
prostate epithelial cells is determined by the specific (Fig. 13.9). Once secreted by the SMC, andromedins
level of prostatic DHT (Kyprianou and lsaacs 1987), diffuse and bind to their cognate receptors on specific
a constant prostatic DHT level is critically required cell types. Within the stromal compartment, such
for the dose-dependent ability of DHT to bind to andromedin binding stimulates myogenesis, denoted

277
Chapter 13: Testosterone and the prostate

Secretory luminal cell 11


Lumen
Secretion (non-proliferative differentiated
compartment; AR positive)
PSA PSA PSA PSA PSA
SFs SFs
HK2 HK2 HK2 HK2 HK2
Transit-amplifying cell 10
12 6 (proliferative precursor
GFs GFs
AR AR AR AR AR compartment; AR negative)
6

Epithelium Epithelial stem cell 9

Stroma
4 Basement membrane
AR AR AR
DFs
GFs/SFs
Smooth muscle cell 3
7

5 5
2 Androgen 8

GFs/SFs Capillary (endothellial cell) 14


2
Mesenchymal stem cell 1
2

Fibroblast 13

GF = Growth factor
Differentiation factor = DF Andromedins
SF = Survival factor
Fig. 13.9 Reciprocal stromal epithelial interactions in the prostate. In the normal prostate, growth and maintenance of prostatic
epithelium depends on paracrine signaling of andromedins (growth and survival factors) produced by supporting stromal cells (smooth
muscle and fibroblasts). Andromedins are secreted due to androgen signaling through AR, a nuclear hormone receptor expressed by prostate
luminal epithelia but not by basal epithelia. Details for the specific numbers are provided in the text.

no. 7 (Fig. 13.9), and vasculogenesis, denoted no. 8 reach its normal adult size by 1820 years of age. This
(Fig. 13.9). Within the epithelial compartment, second prostate growth phase is induced because,
andromedins stimulate epithelium to mature from at puberty, circulating testosterone rises again to a
medullary cords of undifferentiated epithelium which sufficient level to re-stimulate adequate production
contain epithelial stem cells, denoted no. 9 (Fig. 13.9), of stromal andromedins (Fig. 13.3). Such positive
with no canalization, into a simple, stratified, glandu- paracrine stimulatory loops (Fig. 13.9) do not go
lar epithelium composed of a basal layer of cuboidal unopposed, however, since the prostate does not nor-
cells denoted no. 10 (Fig. 13.9), upon which resides a mally continue to grow once it has reached its adult
second layer of columnar secretory luminal cells size, even though there is no decrease in andromedin
denoted no. 11(Fig. 13.9), adjacent to a patent lumen levels. Instead, the prostate reaches its normal adult
(Isaacs 2008). size and then net growth ceases even though circulat-
During the embryonic and early postnatal period, ing levels of androgen are maintained. After reaching
the circulating level of testosterone is sufficient to its adult size, the prostate epithelial compartment
allow the normal stroma to epithelial interaction for enters a steady-state maintenance phase, in which
the initial ontogeny of the prostate. The circulating the rate of epithelial proliferation balances the rate
level of testosterone decreases within the first year of of death such that neither overgrowth nor regression
life, however, such that the prostate does not continue of the gland normally occurs (Litvinov et al. 2003).
to grow (Fig. 13.3). A second phase of prostate growth The realization that both the stromal and epithelial
is initiated during puberty, inducing the prostate to compartments are organized into interactive stem cell

278
Chapter 13: Testosterone and the prostate

units provides a mechanistic understanding of how While an individual prostate stem cell possesses high
the reciprocal positive feedback loops are regulated so self-renewal capacity, it proliferates infrequently to
that the steady-state size of the prostate is maintained renew itself and simultaneously generate progeny for
without overgrowth in the young adult. two distinct cell lineages (Fig. 13.10). The first and
much less frequent lineage commitment is for ter-
13.7 Organization of prostate minal differentiation into a proliferation-quiescent
neuroendocrine cell (NE in Fig. 13.10), which secretes
epithelial and stromal stem cell units a series of peptide growth factors. The second and
Stem cells are defined by their capacity for self- much more common lineage commitment is for
renewal, multi-lineage differentiation and replicative differentiation into a progenitor that undergoes a
quiescence. Pluripotent embryonic stem cells possess limited number of proliferative replications (i.e.
the most plasticity and can give rise to all tissues of amplifications), before transiting a maturation pro-
an organism. During embryogenesis, there is a devel- cess of terminal differentiation (Isaacs 2008). This
opmental process which results in the creation of progenitor is termed a transit-amplifying (TA) cell,
tissue-restricted stem cells, termed adult stem cells, denoted no. 10 in Fig. 13.9. A transit-amplifying cell
which lose their pluripotency, but retain ability to does not express AR protein and is dependent
self-renew and undergo multi-lineage differentiation for proliferation, but not survival, on andromedins
to maintain the tissue. Adult stem cells are generally produced by stromal cells. One of the defining char-
quiescent and reside in a specialized cellular location acteristics of a transit-amplifying cell is its obligatory
known as a niche. The niche provides a microenvir- expression of p63, as well as other basal markers
onment that maintains the balance between quies- such as cytokeratins 5 and 14, Jagged-1 and Notch-1
cence and self-renewal of the stem cell population. (Litvinov et al. 2006).
The concept of adult stem cells in the prostate first An individual transit-amplifying progenitor cell
emerged to explain the profound capacity of this undergoes a limited number of amplifying cell div-
tissue for cyclic regeneration, using male rats as a isions, expanding the cell population derived from
model (Isaacs 1987).These results documented that, a single stem cell, before maturing into an intermedi-
even after 30 rounds of cyclic androgen-ablation- ate cell. This maturation involves downregulation of
induced prostate regression followed by androgen- p63, Jagged-1, Notch-1 and basal cytokeratins 5 and
replacement-induced restoration, which induced 14 expression. A defining characteristic of an inter-
more than 60 additional epithelial cell population mediate cell is its unique expression of PSCA. This
doublings, the prostate epithelial compartment is cell is termed intermediate because it expresses both
completely able to repopulate itself normally (Isaacs luminal lineage-specific cytokeratins 8 and 18, as
1987). This remarkable prostatic regenerative capacity well as the basal lineage-specific cytokeratins 5 and
documents that the self-renewal of stem cells within 15, and AR mRNA, but not AR protein (Litvinov
the adult prostate is androgen independent. et al. 2006). As an intermediate cell migrates upward
Since these original studies 25 years ago, a large from the basal layer to form the luminal layer, it stops
number of independent groups have added to the expressing PSCA and now expresses AR protein.
knowledge of how prostate epithelial stem cell units Engagement of the AR pathway within a luminal cell,
are organized. The results of these combined efforts denoted no. 12 in Fig. 13.9, induces its differentiation
are summarized in Fig. 13.10. Prostate epithelial stem from a cuboidal into a columnar secretory luminal
cells, denoted no. 9 in Fig. 13.9, are present in niches cell expressing prostate specific markers like PSA.
within the basal layer of the epithelial compartment We have recently documented that an additional
at a very low frequency (i.e. 0.51%). A defining function of the ligand-occupied AR in prostatic secre-
characteristic of a prostate epithelial stem cell is that tory luminal cells is to suppress the ability of these
it does not express the AR or p63 proteins (Litvinov secretory cells to proliferate even in the presence of
et al. 2009). This is consistent with the fact that continuously high andromedin levels (unpublished data:
embryonic UGS epithelial cells from either AR or Vander Griend DJ, Litvinov IV, Chen S, Dalrymple SL,
p63 knock-out mice undergo prostatic ontogeny and Antony L, Luo J, DeMarzo AM, Meeker AK, Isaacs JT.
glandular renewal when transplanted in combination Oncogenic addiction of prostate cancer cells to andro-
with wild-type UGS mesenchymal cells (Isaacs 2008). gen receptor involves loss of c-Myc suppression by

279
Chapter 13: Testosterone and the prostate

Basal compartment Luminal compartment


NE

NE

Secretory
luminal
cells

Stem cell Transit amplifying cell Intermediate cells


CASTRATE CASTRATE
Majority of basal cells remain Majority of secretory cells die
Androgen response Independent Dependent
Andromedin response Independent Sensitive Dependent
Selected phenotypic and protien marker expression
Self-renewal capacity Extensive Limited Very limited None
Proliferative index Very low Much higher Low None
Keratin 5 + +
Keratin 14 + +
p63 +

Bcl-2 + + +
P cadherin + ?
GST-pi + +/
C-MET +/ +/ +
p27 Kip1 + /+ +
Keratins 8,18 + +
NKX 3.1 /+ +
PSA /+ +
PSAP /+ +
AR /+ +

Fig. 13.10 Stem cell model of prostatic epithelial cell compartmentalization. The prostate gland consists of a number of stem cell units
that arise from one stem cell. Such a stem cell is located in the basal epithelial layer of the prostate and, upon division, gives rise to a
population of transit-amplifying cells. The latter divide in the basal layer, and a fraction of them differentiate and move into the secretory
luminal epithelial layer. As transit-amplifying cells differentiate and move into a secretory luminal layer from the basal layer, they acquire
expression of a number of genetic markers, as indicated. Symbols: +/, low-level retention of expression by a subset of transit-amplifying
(i.e. intermediate) cells; +, expression of marker; , lack of detectable expression of marker.

androgen receptor/b-Catenin/TCF-4 complexes). and undergo terminal secretory luminal differenti-


These studies document that proliferation of human ation. The mechanism for such androgen-dependent
prostate epithelial cells is dependent upon c-Myc AR-mediated G0 growth arrest is independent of
expression induced by WNT-independent formation Rb, p21, p27, FoxP3, or downregulation of growth
and binding of nuclear b-catenin/TCF-4 complexes factors receptors. Instead, G0 growth arrest involves
to both the 50 and the 30 c-Myc enhancers, thus the formation of AR/b-catenin/TCF-4 complexes
initiating transcription. When prostate epithelial which are unable to bind the 50 /30 enhancers and
cells expressing AR are exposed to androgen, c-Myc initiate c-Myc transcription. Such binding to b-catenin/
expression is suppressed and the cells arrest in G0 TCF-4 complexes does not occur when AR is mutated

280
Chapter 13: Testosterone and the prostate

in its DNA-binding domain. The causal importance (Fig. 13.9), including insulin-like growth factor 1
of this mechanism is confirmed by the ability of (IGF-1), FGF-7 and-10, and vascular endothelial
constitutive c-Myc expression to prevent androgen- growth factor (VEGF) (Isaacs 2008).
dependent growth arrest of AR-expressing prostate Insulin-like growth factor 1 is not only a para-
epithelial cells. It is this ligand-occupied AR suppres- crine factor for prostate epithelial cells but also a
sion of proliferation of the secretory luminal cells critical autocrine factor, denoted no. 7 in Fig. 13.9,
which prevents the androgen-stimulated stroma to for prostate stromal cells (Isaacs 2008). Another
epithelial positive feed-forward loop (Fig. 13.9) from androgen-dependent paracrine factor produced by
inducing continuous net prostatic growth in the pres- SMC, denoted no. 8 (Fig. 13.9), is VEGF. Vascular
ence of a continuous supply of andromedins. endothelial growth factor maintains the endo-
Since secretory luminal cells are the terminal stage thelial cells, denoted no. 14 (Fig. 13.9), and thus the
of maturation of a hierarchical expanding popula- blood supply in the prostate stromal compartment
tion of cells comprising an individual stem cell unit affecting both the stromal and epithelial cells (Joseph
(Fig. 13.10), these secretory luminal cells are quanti- et al. 1997).
tatively the major epithelial phenotype present in the
gland even though they are proliferatively quiescent
(i.e. terminally differentiated). Unlike their prolifer- 13.8 Androgen in benign prostatic
ating precursors, terminally differentiated secretory hyperplasia and the clinical use
luminal cells, denoted no. 11 in Fig. 13.9, acquire a
dependence on stromally derived andromedins for of SRD5A inhibitors for its treatment
their survival. Hence, androgen deprivation induces The realization that prostate is organized in to stem
apoptosis of these secretory luminal cells due to a cell units provides a framework for understanding the
decrease in andromedin levels. It is the death of these mechanism for preventing either the regression or
secretory luminal cells which predominately accounts overgrowth of the normal adult gland. At about the
for the regression of the prostate induced by andro- age of 40, however, in the vast majority of men, this
gen deprivation. kinetic balance is disrupted and, with further aging,
Besides prostate epithelial stem cells, there are the gland enlarges and eventually produces BPH
also mesenchymal stem cells within prostate stroma. (Rhodes et al. 1999). In BPH, there is an increase in
This conclusion is based upon the demonstration the cellular content of the transition zone of the
that there is a self-renewing subpopulation of prostate prostate (Fig 13.3). This neoplastic growth involves:
stromal cells from BPH tissue which express: (1) (1) an enhanced number of epithelial and stromal
mesenchymal stem cell markers; (2) strong prolifera- stem cell units; (2) an enhanced number of prolifera-
tive potential; and (3) ability to differentiate in to tions by transit-amplifying cells before these mature
fibroblastic, myogenic, adipogenic and osteogenic into non-proliferating luminal secretory cells; and/or
lineages (Lin et al. 2007). Of these potential lineages, (3) decreased ability of AR to limit the proliferation
the most characteristic commitment of progeny from of luminal secretory cells. Benign prostatic hyperpla-
prostate mesenchymal stem cell is to differentiate sia characteristically is associated with an enhanced
into a fibroblast, denoted no. 13, or a smooth muscle number of stromal cells. Since at least a subset of
cell, denoted no. 3 (Fig. 13.9). This latter commit- these stromal cells express AR and thus andromedins,
ment, as discussed earlier, is due to embryonic para- androgen regulation within these stromal cells may
crine effects of prostate epithelial cells, denoted no. 4 be abnormal, leading to enhanced andromedin pro-
in Fig. 13.9, which permanently commit the mesenchy- duction. Theoretically, to inhibit such enhanced
mal stem cells to give rise to a subset of progeny andromedin production, androgen ablation could be
which can mature into AR-expressing SMC (Cunha utilized to treat BPH. Unfortunately, such systemic
et al. 1996). In the adult, the maturation of these androgen ablation has other unacceptable side-effects
SMC requires paracrine factors secreted by prostate on bone density, muscle mass and libido. For these
epithelial cells, resulting in their expression of both reasons, BPH is often treated medically with SRD5A
5a-reductase and AR proteins (Bayne et al. 1998). inhibitors, since they decrease prostate tissues levels
Once these SMC mature, androgen binding to AR of DHT without lowering the systemic level of testos-
induces their secretion of andromedins, denoted no. 6 terone (Nasland and Miner 2007).

281
Chapter 13: Testosterone and the prostate

F3C Fig. 13.11 Chemical structure of the


two clinically approved SRD5A inhibitors,
finasteride and dutasteride.
HN CF3
O O NH

H
H
H H H
H
O N O N
H H H H

Finasteride Dutasteride

GSTP1 promoter methylation


Telomere shortening
Engagement of AR in a novel way Telomerase activation
Oxidative stress Beginning accumulation of genomic Continued accumulation of
Cell injury and regeneration changes genomic changes
(e.g. loss of 8p) (e.g. 8p24 gain)

Proliferative
Normal inflammatory PIN
atrophy (PIA)
Invasive
carcinoma
Secretory
Basal

Inflammation, dietary factors, inherited factors


Fig. 13.12 Overview of prostatic carcinogenesis. PIN = prostatic intraepithelial neoplasia.

There are two clinically approved, orally active, the BPH-enlarged prostate by 25%, without lowering
small molecule SRD5A inhibitors: finasteride and serum testosterone and thus minimizing androgen-
dutasteride (Fig. 13.11). Finasteride is the older of ablation side-effects (Foley and Kirby 2003).
the two approved drugs, and it is a competitive inhibi-
tor of the human SRD5A1 isozyme with a Ki = 360 
40 nM, and an irreversible inhibitor of the human 13.9 Role of androgen in prostate
SRD5A2 isozyme with a Ki of 69  1 nM (Xu et al.
2006). Due to its rapid serum half-life, finasteride is carcinogenesis
a SRD5A2 selective drug (Xu et al. 2006). Like finas- Prostate carcinogenesis is the result of the accumulation
teride, dutasteride is also a competitive inhibitor of of multiple genetic and epigenetic changes, whereby
human SRD5A1 with a Ki of 6  1 nM, and an chronic and acute inflammation, in conjunction with
irreversible inhibitor of the human SRD5A2 isozyme dietary and other environmental factors, targets pro-
with a Ki of 7  3 nM (Xu et al. 2006). Due to its state epithelial cells for injury and destruction (De
extended serum half-life (i.e. >3 days), dutasteride is Marzo et al. 2007; Nelson et al. 2007; Fig. 13.12). The
a dual SRD5A1 and 2 inhibitor (Xu et al. 2006). precursor for most peripheral zone prostatic carcin-
Chronic daily treatment with either of these SRD5A omas is thought to be high-grade prostatic intraepithe-
inhibitors lowers prostatic DHT, which decreases stro- lial neoplasia (HGPIN) (McNeal and Bostwick 1986).
mal andromedin production, thus reducing the size of It is believed that HGPIN arises from low-grade PIN.

282
Chapter 13: Testosterone and the prostate

The cell type of origin for HGPIN is incompletely measure. Although elevated in many of the cells in
understood. A widely held view of carcinogenesis is PIA, GSTP1 expression is eventually lost in some cells
that the common carcinomas generally arise in self- as the result of aberrant methylation of the CpG
renewing tissues in which dividing cells acquire som- islands of the GSTP1 gene promoter (Lin et al.
atic genetic alterations in growth-regulatory genes. In 2001). Indeed, such aberrant methylation of the
normal human prostate epithelium, cell division takes GSTP1 promoter is one of the earliest molecular
place almost exclusively in the basal cell compartment abnormalities characteristic of prostate cancer cells.
where the tissue stem and presumably the transit- This heritable epigenetic alteration places these cells
amplifying cells reside (Bonkhoff et al. 1994; Bonkh- at increased risk for the accumulation of additional
off 1998). The secretory luminal cells do not normally genetic damage, with acceleration of the neoplastic
proliferate and are the terminally differentiated cells process toward PIN (Lin et al. 2001). One of these
that perform the androgen-regulated differentiated additional genetic changes involves telomerase
functions of the prostate, such as PSA production shortening by PIN cells. This appears to increase their
and secretion. Both prostate cancer and HGPIN cells genetic instability, driving further genetic damage
possess many phenotypic and morphological features and producing invasive cancers (Meeker et al. 2002;
of secretory luminal cells (i.e. cytokeratin 8 and 18, Fig. 13.12).
PSA, HK, PSMA and AR expression); yet they also During the initiation of prostate carcinogenesis,
contain features of the basal transit-amplifying cell there are distinct hard wiring changes in the AR
compartment such as c-MET expression, DNA repli- signaling pathways. Normally the proliferating tran-
cation and extensive self-renewal (Verhagen et al. sit-amplifying cells in the basal epithelial layer do not
1992; De Marzo et al. 1998a; Meeker et al. 2002; van express the androgen receptor, or express only low
Leenders et al. 2002). Thus, in carcinoma these stem levels of AR. As discussed, during their maturation,
cell and transit-amplifying celllike features have been these cells eventually express higher levels of AR.
shifted up from the basal into the secretory luminal Once a critical AR level is reached, the occupancy of
compartment (De Marzo et al. 1998b; Meeker et al. AR with ligand inhibits proliferation of these cells and
2002). Based upon these facts, it has been postulated induces their differentiation into secretory luminal
that the cell of origin for prostate cancer is an inter- cells. In contrast, the intermediate type of prolifer-
mediate, prostatic epithelial cell, presumably derived ating cells in PIA variably expresses higher levels of
from the basal transit-amplifying population, which AR, and such AR expression is further enhanced in
undergoes the initial malignant molecular changes proliferating cells in HGPIN (De Marzo et al. 2001).
allowing gene expression and morphological features Associated with this enhanced expression of the AR is
of both basal and secretory luminal cells (Verhagen the decreased expression of ERb by HGPIN cells
et al. 1992; De Marzo et al. 1998a; De Marzo et al. (Fixemer et al. 2003). These results document that
1998b; Meeker et al. 2002; van Leenders et al. 2002; hard wiring changes occur in the AR/ERb signaling
Vander Griend et al. 2008). pathways even at this early stage of cancer develop-
This conclusion is also supported by the demon- ment, since now AR-expressing/ERb-negative cells
stration that increased proliferation occurs as a regen- are proliferating and not growth arrested.
erative response in cells with a transit-amplifying or These changes produce gain of function ability
intermediate phenotype (Meeker et al. 2002; van by AR so that it now engages the molecular signaling
Leenders et al. 2002). The site of these phenotypically pathways directly, stimulating the proliferation and
intermediate, initiated cells appears not to be random survival of these initiated prostatic cells (Vander
within the prostate. Instead, they are enriched in sites Griend et al. 2010). Unlike the paracrine situation in
of focal glandular atrophy where the luminal epithe- the normal prostate, in which such growth regulation
lial cells, atrophic in appearance, are quite prolifera- is initiated by AR binding to genomic sequences in the
tive and often surrounded by inflammation within the nuclei of stromal cells, during prostatic carcinogenesis
gland. Therefore, these sites have been termed pro- genomic AR binding within the transformed cells
liferative inflammatory atrophy (PIA) (De Marzo itself activates this growth regulation. Due to these
et al. 1999; Fig. 13.12). In this process, glutathione hard wiring changes, there is a conversion from
S-transferase pi 1 (GSTP1) expression is elevated in paracrine to a cell-autonomous autocrine AR signal-
many of the cells in PIA as a genome-protective ing pathway in invasive prostate cancer (Gao and

283
Chapter 13: Testosterone and the prostate

LHRH
Hypothalamus

Progesterone, estrogen,
androgen receptor
binding and signaling
LHRH Antiandrogens
agonists
LHRH antagonists
LHRH receptor
Hypothalamus
(Prolonged
exposure)
LHRH
Pituitary
LH

LHRH-R

Brain LH receptor
LH Pituitary

Castration Testis
Abiraterone
Testosterone Ketoconazole
LH-R 5-
Adrenal
Testosterone Reductase DHT
glands Cholesterol Androgen
Cholesterol Androgens
AR
Testosterone DHT AR binding
Nucleus and
Growth and activation of
Testosterone survival genes
Androgens 5-Reductase target genes
inhibitors
Prostate Antiandrogens
Testis Adrenal gland Prostate
Fig. 13.13 Overview of the hypothalamic-pituitary-testicular axis and sites of therapeutic disruption by chemical agents.

lsaacs 1998; Gao et al. 2001; Vander Griend et al. into the blood results in inhibition of testosterone
2010). These gain of function hard wiring changes production and secretion by the testes, producing
pathologically allow androgen/AR complexes to bind a medical castration (Denmeade and Isaacs 2002;
to and enhance expression of survival and prolifer- Fig. 13.13). These drugs rarely actually cure the cancer,
ation genes which physiologically are not affected by however, because the initial response is almost always
these complexes in either normal transit-amplifying followed by a relapse to a castration-resistant stage.
or secretory luminal cells (Gao and lsaacs 1998; Gao The development of the castration-resistant state
et al. 2001; Vander Griend et al. 2010). is not commonly associated with the loss of AR
expression (Hobisch et al. 1996; Taplin et al. 1999;
13.10 Androgen ablation as therapy Linja et al. 2001; Shah et al. 2004). In fact, enhanced
expression of AR is the most common phenotypic
for metastatic prostate cancer change characteristically associated with the develop-
Due to the fact that, during prostate carcinogenesis, ment of castration-resistant prostate cancer (Chen
AR is converted from a growth suppressor to an et al. 2004; Taylor et al. 2010). While less than 10%
oncogene promoting cancer cell proliferation, patients of these cancers have undergone AR mutations
with metastatic prostate cancer initially receive long- (Taplin et al. 2003), approximately 30% of such cas-
acting super-agonists of luteinizing hormone-releasing tration-resistant prostatic cancers over-express AR
hormone (LHRH) which downregulate the cell surface due to amplification in the gene (Brown et al. 2002;
expression of LHRH receptors on gonadotrophs in the Hyytinen et al. 2002; Taylor et al. 2010). A growing
anterior pituitary; thus preventing release of LH from body of additional evidence points to the accumula-
these cells (Fig. 13.13). Downregulation of LH release tion of molecular changes that reduce the threshold of

284
Chapter 13: Testosterone and the prostate

N (B)
(A) O F
N
O
O MeHN S
S N CF3
H N CF3
O OH N

F O
Bicalutamide MDV3100

N N

(C) N (D)

O O
H
N N O O H H
CI
H HO
CI
Ketoconazole Abiraterone
Fig. 13.14 Chemical structure of agents targeted at disrupting androgenic stimulation of prostate cancer cells. A=bicalutamide (Casodex);
B=MDV3100; C=ketoconazole; and D=abiraterone.

AR ligand required for proliferation and survival Despite encouraging clinical results with next gener-
(Isaacs and Isaacs 2004). Thus the AR is still a thera- ation drugs (e.g. MDV3100 and abiraterone) targeted
peutic target in castration-resistant prostate cancer. at inhibiting the residual AR signaling in patients with
In an attempt to block the residual AR signaling CRPC, responses are variable and short-lived (Evans
in the castration-resistant state, competitive AR et al. 2011; de Bono et al. 2011).
antagonists (i.e. antiandrogens, like bicalutamide
(Casodex; Fig. 13.14A) and, in more recent phase II 13.11 SRD5A inhibitors for the
clinical trials, the novel AR antagonist MDV3100
(Fig. 13.14B; Scher et al. 2010)) are often combined prevention of prostate cancer
with LHRH super-agonists (Evans et al. 2011). Unfor- The realization that AR signaling is converted from a
tunately, such additional androgen-ablation therapies suppressor to an oncogenic stimulator of epithelial
are also usually not curative (Evans et al. 2011). To cell proliferation early in prostate carcinogenesis has
more fully suppress circulating androgen production, raised the issue of whether prostate cancer develop-
particularly that derived from both the adrenals as ment can be prevented by reducing AR signaling.
well as testes within such castrate-resistant metastatic Based upon their safe use in the treatment of BPH,
patients, LHRH super-agonists have been combined two large RCTs, the Prostate Cancer Prevention Trial
with inhibitors of the cytochrome P450 steroid (PCPT) (Thompson et al. 2003) and the Reduction
17a-mono-oxygenase enzyme (i.e. CYP17A1 aka by Dutasteride of Prostate Cancer Events (REDUCE)
17a-hydroxylase/17,20 lyase), which converts pregne- trial (Andriole et al. 2010) evaluated daily use of
nolone/progesterone into DHEA and androstene- finasteride 5 mg versus placebo for seven years and
dione, since these intermediates must be formed for daily use of dutasteride 0.5 mg versus placebo for four
production of testosterone in both glands (Fig. 13.3). years, respectively, for the reduction in the risk of
In clinical trials, either the non-selective inhibitor, prostate cancer in men at least 50 years of age. The
ketoconazole (Fig. 13.14C; Keizman et al. 2012) or trials demonstrated an overall 25% reduction in pro-
the more specific inhibitor, abiraterone (Fig. 13.14D; state cancer diagnoses with finasteride 5 mg and
de Bono et al. 2011) have been used based upon the dutasteride treatment. This overall reduction was due
idea that castration-resistant prostate cancer (CRPC) to a decreased incidence of lower risk (i.e. Gleason
remains hormone driven, with intratumoral steroid Score 6) prostate cancers. However, both trials showed
synthesis fueling tumor growth (Cai et al. 2011). an increased incidence of high-grade (i.e. Gleason

285
Chapter 13: Testosterone and the prostate

Score >7) prostate cancer with finasteride and dutaste- to environmental conditions. The goal is to determine
ride treatment. Based upon an expert panel review of if a clinical response can be achieved through this non-
these results in June of 2011, the FDA informed health- adaptive rapid cycling approach in men with CRPC.
care professionals that the Warnings and Precautions
section of the labels for the SRD5A inhibitor class of
drugs had to be revised to include new safety infor- 13.13 Side-effects of androgen
mation about the increased risk of being diagnosed replacement/ablation in the
with a more serious form of prostate cancer (high-
grade prostate cancer). This risk appears to be low, aging male
but healthcare professionals should be aware of this Besides its effects upon normal and abnormal growth
safety information, and weigh the known benefits and physiology of the prostate, testosterone is also
against the potential risks when deciding to start or critical for maintenance of bone and muscle metabol-
continue treatment with SRD5A inhibitors in men. ism, as well as libido. For this reason, aging males
who have an insufficient level of serum testosterone
(i.e. hypogonadal males) suffer clinically from loss
13.12 New direction for treatment of bone and muscle mass as well as a decreased libido.
of castration-resistant prostate cancer: Such aging hypogonadal males are candidates for
exogenous testosterone replacement. Due to its
bipolar androgen therapy growth-promoting effects on the prostate, however,
Androgen ablation is highly effective palliative ther- such hormonal replacement therapy could enhance
apy for metastatic prostate cancer, but eventually all the development of BPH and/or prostate cancer. Thus,
men relapse. The demonstration that AR expression the decision to initiate such replacement hormonal
continues in androgen-ablated patients has resulted therapy must be evaluated on risk vs. benefit analysis
in the classification castration-resistant prostate for each patient individually (see Chapter 16).
cancer (CRPC), and has led to the development of The side-effects observed in naturally developed
new second-line agents targeted at reducing residual hypogonadal males are also a problem in males either
AR signaling. Paradoxical to these second-line at high risk of developing prostate cancer that are
approaches is the observation that the growth of treated with androgen-ablation therapy as a preventa-
AR-expressing human CRPC can be inhibited by tive modality, or in patients with clinically established
supraphysiological levels of androgens. This response prostate cancer who are being given androgen-
appears to be due to high-dose androgen inhibiting ablation as therapy. In order to allow replacement
re-licensing of DNA in CRPC cells expressing high hormonal therapy in hypogonadal patients and lessen
levels of AR (Litvinov et al. 2006; Vander Griend et al. side-effects of such testosterone-ablative therapies
2007; DAntonio et al. 2009; Denmeade and Isaacs in patients with established prostate cancer, small
2010). It may also be due to recently described effects molecule SARMs are being developed which retain
of androgen in inducing double strand DNA breaks the positive androgenic effects on bone, muscle and
(Haffner et al. 2010). Based on available preclinical libido, but which have little or no growth-stimulatory
data demonstrating the effects of supraphysiological effects on the prostate (Bhasin and Jasuja 2009). This
levels of testosterone on inhibition of growth of approach is possible due to the increasing basic know-
CRPC xenografts, we have initiated a clinical trial in ledge about the mechanism(s) of such androgenic
men with CRPC testing the effect of monthly treat- effects at the molecular level (Litvinov et al. 2003).
ments with an im depot injection of testosterone. This These molecular studies have documented that the
im formulation achieves supraphysiological levels of binding of natural and synthetic SARMs induces a
testosterone that cannot be achieved with standard spectrum of conformational changes in the androgen
testosterone gel-based applications. The supraphysio- receptor. This spectrum of conformations results in
logical testosterone level is followed by a rapid drop differential ability of the SARM-occupied AR to
to castrate levels of testosterone with each cycle of dimerize and bind to specific target genes and specific
therapy (Denmeade and Isaacs 2010). This bipolar transcriptional cofactors, inducing either stimulation
androgen therapy does not allow time for prostate or repression of transcription. Thus, the development
cancer cells to adapt their AR expression in response of such SARMs will usher in an exciting time during

286
Chapter 13: Testosterone and the prostate

which clinical testing will determine whether modu-


lating testosterones effects will have an impact on growth arrest by downregulating c-Myc
expression.
the prevention and treatment of multiple diseases of
 Prostate cancers are derived from epithelial cells.
the aging male.
During prostatic carcinogenesis, molecular
changes induce a conversion from a paracrine to
13.14 Key messages an autocrine pathway so that the AR then directly
 The prostate is the most common site stimulates the proliferation and survival of
of neoplastic transformation in the human body. prostate cancer cells.
 There are two types of prostatic neoplasms:  Therapies which lower androgen levels or
one benign (i.e. BPH); the other malignant block AR signaling have both preventative
(i.e. prostate carcinoma). and treatment effect on prostatic cancer and
 Testosterone is the major growth and functional BPH.
regulator of both the normal and abnormal  Besides its effects upon normal and
prostate. abnormal growth and physiology of the
 The prostate epithelium which gives rise to prostate, testosterone is also a critical regulator
prostate cancer is organized functionally in of bone and muscle metabolism, as well as
stem cell units composed of stem cells, libido. Therefore, therapies which reduce
transit-amplifying cells, intermediate cells and testosterones effects on the development and
secretory luminal cells. clinical progression of either BPH or prostate
 Testosterone is a prohormone in the prostate, cancer have major side-effects upon quality
where it is metabolized to both a more potent of life.
androgen (i.e. DHT) and to an estrogenic  In addition, there are aging males who suffer
metabolite (i.e. 3b-diol). from abnormally low serum testosterone levels
 Dihydrotestosterone binds to the AR within with similar quality-of-life side-effects. These
prostatic stromal cells to induce the production patients can be supplemented with exogenous
and secretion of paracrine growth factors known testosterone, but they must be carefully
as andromedins. monitored for development of BPH and prostate
 Andromedins diffuse into the epithelial cancer.
compartment where they stimulate the  To allow replacement therapy in patients with
proliferation of transit-amplifying cells and the low serum testosterone and lessen side-effects
survival of transit-amplifying, intermediate and of testosterone, ablative therapies in
secretory luminal cells (i.e. paracrine androgen patients with established prostate cancer,
axis in normal prostate). small molecule selective androgen response
 Dihydrotestosterone binds to the AR in secretory modifiers (SARMs) are being developed, which
luminal cells and directly induces the retain the positive androgenic effects on
transcription of prostate differentiation markers, bone, muscle and libido, but which have little
such as PSA, HK2 and prostatic-specific acid or no growth stimulatory effects on the
phosphatase, and induces their terminal prostate.

13.15 References dutasteride on the risk of


prostate cancer. N Engl J Med
Berry SI, Coffey DS, Walsh PC,
Ewing LL (1984) The development
Andersson S, Russell DW (1990) 362:11921202 of human benign prostatic
Structural and biochemical hyperplasia with age. J Urol
properties of cloned and expressed Bayne CW, Donnelly F,
Chapman K, Bollina P, Buck C, 132:474479
human and rat steroid 5a-
reductases. Proc Natl Acad Sci USA Habib F (1998) A novel coculture Bhasin S, Jasuja R (2009) Selective
87:36403644 model for benign prostatic androgen receptor modulators as
hyperplasia expressing both function promoting therapies.
Andriole GL, Bostwick DG, Brawley
isoforms of 5alpha-reductase. Curr Opin Clin Nutr Metab Care
OW, Gomella LG, Marberger M,
J Endocrinol Metab 83:206213 12:232240
Montorsi F, Pettaway CA,
Tammela TL, Teloken C, Tindall Berman DM, Russell DW (1993) Biswas MC, Russell DW (1997)
DJ, Somerville MC, Wilson TH, Cell-type-specific expression of rat 5 Expression cloning and
Fowler IL, Rittmaster RS; REDUCE alpha-reductase isozymes. Proc Natl characterization of oxidative
Study Group (2010) Effect of Acad Sci USA 90:93599363 17b- and 3a-hydroxysteroid

287
Chapter 13: Testosterone and the prostate

dehyrogenases from rat and antiandrogen therapy. Nat Med inflammatory atrophy of the
human prostate. J Biol Chem 10:3339 prostate: implications for prostatic
272:1595915966 Coffey DS (1992) The molecular carcinogenesis. Am J Pathol
Bonkhoff H (1998) Neuroendocrine biology endocrinology and 155:19851992
cells in benign and malignant physiology of the prostate and De Marzo AM, Putzi NIT, Nelson WG
prostate tissue: morphogenesis seminal vesicles. In: Walsh PC, (2001) New concepts in the
proliferation and androgen receptor Retik AB, Stamey TA, Vaughan EL pathology of prostatic epithelial
status. Prostate Suppl 8:1822 (eds) Campbells Textbook of carcinogenesis Urology 57
Urology. Saunders, Philadelphia, pp (Suppl 1):103114
Bonkhoff H, Stein U, Remberger K
221266
(1994) The proliferative function of De Marzo AM, Platz EA, Sutcliffe S,
basal cells in the normal and Cunha GR, Hayward SW, Daihiya R, Xu J, Gronberg H, Drake CG, Nakai
hyperplastic human prostate. Foster BA (1996) Smooth muscle Y, Isaacs WB, Nelson WG (2007)
Prostate 24:114118 epithelial interactions in normal Inflammation in prostate
and neoplastic prostate carcinogenesis. Nat Rev Cancer
Brown RS, Edwards J, Dogan A, Payne H,
development. Acta Anat 155:6372 7:256269
Harland SJ, Bartlett TM, Masters JR
(2002) Amplification of the DAntonio JM, Vander Griend DJ, Denmeade SR, Isaacs JT (2002)
androgen receptor gene in bone Isaacs JT (2009) DNA licensing as a A history of prostate cancer
metastases from hormone- novel androgen receptor mediated treatment Nat Rev Cancer 2:389396
refractory prostate cancer. Pathol therapeutic target for prostate
Denmeade SR, Isaacs JT (2010) Bipolar
98:237244 cancer. Endocr Relat Cancer
16:325332 androgen therapy: the rationale for
Bruchovsky N, Wilson JD (1968) The rapid cycling of supraphysiologic
conversion of testosterone in 5a- de Bono JS, Logothetis CJ, Molina A, androgen/ablation in men with
androstan-17b-ol-3one by the rat Fizazi K, North S, Chu L, Chi KN, castration resistant prostate cancer.
prostate in vivo and in vitro. J Biol Jones RJ, Goodman OB Jr, Saad F, Prostate 70:16001607
Chern 243:20122021 Staffurth JN, Mainwaring P,
Harland S, Flaig TW, Hutson TE, Eick GN, Thornton JW (2011)
Cai C, Chen S, Ng P, Bubley GJ, Nelson Cheng T, Patterson H, Hainsworth Evolution of steroid receptors from
PS, Mostaghel EA, Marck B, an estrogen-sensitive ancestral
JD, Ryan CJ, Sternberg CN, Ellard
Matsumoto AM, Simon N, Wang SL, Flchon A, Saleh M, Scholz M, receptor. Mol Cell Endocr 334:3138
H, Chen S, Balk SP (2011)
Efstathiou E, Zivi A, Bianchini D, Evans MJ, Smith-Jones PM, Wongvipat J,
Intratumoral de novo steroid Loriot Y, Chieffo N, Kheoh T, Haqq
synthesis activates androgen Navarro V, Kim S, Bander NH,
CM, Scher HI; COU-AA-301 Larson SM, Sawyers CL (2011)
receptor in castration-resistant Investigators (2011) Abiraterone
prostate cancer and is upregulated Noninvasive measurement of
and increased survival in metastatic androgen receptor signaling with a
by treatment with CYP17A1 prostate cancer. N Engl J Med
inhibitors. Cancer Res 71:65036513 positron-emitting
364:19952005 radiopharmaceutical that targets
Cantagrel V, Lefeber DJ, Ng BG, Guan Dehm SM, Tindall DJ (2007) Androgen prostate-specific membrane antigen.
Z, Silhavy JL, Bielas SL, Lehle L, receptor structural and functional Proc Natl Acad Sci USA
Hombauer H, Adamowicz M, elements: role and regulation in 108:95789582
Swiezewska E, DE Brouwer AP, prostate cancer. Mol Endocrinol
Blumel P, Sykut-Cegielska J, Fixemer F, Remberger K, Bonkhoff H
21:28552863 (2003) Differential expression of the
Houliston S, Swistun D, Ali BR,
Dobyns WB, Babovic-Vuksanovic D, De Marzo AM, Meeker AK, Epstein JI, estrogen receptor beta (ERbeta) in
van Bokhoven H, Wevers RA, Coffey DS (1998a) Prostate stem cell human prostate tissue,
Raetz CR, Freeze HH, Morava E, compartments: expression of the premalignant changes, and in
Al-Gazali L, Gleeson JG (2010) cell cycle inhibitor p27kip I in primary, metastatic, and recurrent
SRD5A3 is required for converting normal hyperplastic and neoplastic prostatic adenocarcinoma. Prostate
polyprenol to dolichol and is cells. Am Pathol 153:911919 54:5987
mutated in a congenital De Marzo AM, Nelson WG, Meeker Foley CL, Kirby KS (2003) 5
glycosylation disorder. Cell AK, Coffey DS (1998b) Stem cell alpha-reductase inhibitors: whats
142:203217 features of benign and malignant new? Curr Opin Urol 13:3137
Chen CD, Welsbie DS, Tran C, Baek prostate epithelial cells. J Urol Frasier SD, Gafford F, Horton RD
SH, Chen R, Vessella R, Rosenfeld 160:23812392 (1969) Plasma androgens in
M, Sawyers CL (2004) Molecular De Marzo AM, Marchi VL, Epstein JI, childhood and adolescence. J CLin
determinants of resistance to Nelson WG (1999) Proliferative Endocrinol Metab 29:14041408

288
Chapter 13: Testosterone and the prostate

Gao J, lsaacs JT (1998) Development of Hyytinen ER, Haapala K, Thompson I, than one human steroid 5a-
an androgen receptor null model for Lappalainen I, Roiha M, Rantala I, reductase. J Clin Invest 89:293300
identifying the site of initiation for Helin HJ, Janne OA, Vihinen NI, Joseph IB, Nelson JB, Denmeade SR,
androgen stimulation of Palvimo JJ, Koivisto PA (2002) Isaacs JT (1997) Androgens regulate
proliferation and suppression of Pattern of somatic androgen vascular endothelial growth factor
programmed (apoptotic) death of receptor gene mutations in patients content in normal and malignant
PC-82 human prostate cancer cells. with hormone-refractory prostate prostatic tissue. Clin Cancer
Cancer Res 58:32993306 cancer. Lab Invest 82:13911598 Res 3:25072511
Gao J, Arnold JT, lsaacs JT (2001) Isaacs JT (1984) Common Keizman D, Huang P, Carducci MA,
Conversion from a paracrine to characteristics of human and canine Eisenberger MA (2012)
an autocrine mechanism of benign prostatic hyperplasia. In: Contemporary experience with
androgen-stimulated growth during Kimball FA (ed) New Approaches to ketoconazole in patients with
malignant transformation of the Study of Benign Prostatic metastatic castration-resistant
prostatic epithelial cells. Cancer Hyperplasia. AR Liss, New York, prostate cancer: clinical factors
Res 61:50385044 pp 217234 associated with PSA response and
Godoy A, Kawinski E, Li Y, Oka D, Isaacs JT (1987) Control of cell disease progression. Prostate
Alexiev B, Azzouni F, Titus MA, proliferation and cell death in the 72:461467
Mohler J (2011) 5a reductase type 3 normal and neoplastic prostate: a Kyprianou N, Isaacs JT (1987) Quantal
expression in human benign and stem cell model. In: Rogers CH, relationship between prostatic
malignant tissues: a comparative Coffey DS, Cunha G, Grayhack J, dihydrotestosterone and prostatic
analysis during prostate cancer Hinman F, Horton R (eds) Benign cell content: critical threshold
progression. Prostate 71:10331046 Prostatic Hyperplasia. Report No. concept. Prostate 11:4150
Gorski RA (2002) Hypothalamic INH 872881. Department of
Health and Human Services, Kyprianou N, Isaacs JT (1988)
imprinting by gonadal steroid Activation of programmed cell
hormones. Adv Exp Med Biol National Institutes of Health,
Bethesda, MD, pp 8594 death in the rat ventral prostate
11:5770 after castration. Endocrinology
Gottlieb B, Vasiliou DM, Lumbroso R, Isaacs JT (2008) Prostate stem cells and 122:552562
Beitel LK, Pinsky L, Trifiro MA benign prostatic hyperplasia.
Prostate 68:10251034 Labrie F, Sugimoto Y, Luu-The V,
(1999) Analysis of exon 1 mutations Simard J, Lachance Y, Bachuarov D,
in the androgen receptor gene. Isaacs JT, Isaacs WB (2004) Androgen Leblanc G, Durocher F, Paquet N
Hum Mutat 14:527539 receptor outwits prostate cancer (1992) Structure of human type II
Grumbach MM (2002) The drugs. Nat Med 10:2627 5a-reductase gene. Endocrinology
neuroendocrinology of human Isaacs JT, McDermott IR, Coffey DS 131:15711573
puberty revisited. Horm Res 57 (1979) The identification and Liao S, Leong T, Tymocyko JL (1972)
(Suppl 2):214 characterization of a new C19O3 Structural recognition in
Haffner MC, Aryee MJ, Toubaji A, steroid metabolite in the rat ventral interactions of androgens and
Esopi DM, Albadine R, Gurel B, prostate: 5a-androstane- receptor problems and in their
Isaacs WB, Bova GS, Liu W, Xu J, 3b,6a,17b-triol. Steroids 33:639675 association with nuclear
Meeker AK, Netto G, De Marzo Jemal A, Bray F, Center MM, Ferlay components. J Steroid Biochem
AM, Nelson WG, Yegnasubramanian ME, Ward E, Forman D (2011) 3:401407
S (2010) Androgen-induced Global cancer statistics. CA Cancer Lin VK, Wang SY, Vasquez DV, Xu
TOP2B mediated double strand J Clin 61:6990 CC, Zhang S, Tang L (2007)
breaks and prostate cancer Jenkins EP, Hsieh C-L, Milatovich A, Prostatic stromal cells derived from
gene rearrangement. Nat Genet Norminton K, Berman DM, benign prostatic hyperplasia
42:668675 Franke U, Russel PW (1991) specimens posses stem cell like
Hobisch A, Culig Z, Radnayr C, Characterization and chromosomal property. Prostate 67:12651276
Bartsch C, Klocker H, Hittinair A mapping of a human steroid 5a- Lin X, Tascilar M, Lee WH, Vles WJ,
(1996) Androgen receptor status of reductase gene and pseudogene and Lee BH, Veerawamy R, Asgari K,
lymph node metastases from mapping of the mouse homologue. Freije D, van Rees B, Gage WR,
prostate cancer: Prostate Genomics 11:11021112 Bova GS, Isaacs WB, Brooks JD, de
28:129135 Jenkins EP, Anderson S, Imperato- Weese TL, De Marzo AM, Nelson
Huggins C, Johnson MA (1947) McGinley J, Wilson JD, Russell WG (2001) GSTPI CpG island
Carcinoma of the bladder and DW (1992) Genetic and hypermethylation is responsible for
prostate. JAMA 135:11461152 pharmacologic evidence for more the absence of GSTPI expression in

289
Chapter 13: Testosterone and the prostate

human prostate cancer cells. Am occurrence and experimental Rhodes T, Girman CJ, Jacobsen SJ,
J Pathol 159:18151826 production in animals. Surgery Roberts RO, Guess HA, Lieber MM
Linja MJ, Savinainen KJ, Sakamaki OR, 16:152167 (1999) Longitudinal prostate growth
Tammela TLJ, Vessella RL, Nasland MJ, Miner M (2007) A review rates during 5 years in randomly
Visakorpi T (2001) Amplification of the clinical efficacy and safety selected community men 40 to 79
and overexpression of androgen of 5alpha-reductase inhibitors for years old. J Urol 161:11741179
receptor gene in hormone- the enlarged prostate. Clin Ther Rizner TL, Lin HK, Peehl DM,
refractory prostate cancer. Cancer 29:1725 Steckelbroeck S, Bauman DR,
Res 61:35503555 Nelson WG, Yegnasubramanian S, Penning TM (2003) Human type 3
Agoston AT, Bastian PJ, Lee BH, 3a-hydroxysteroid dehydrogenase
Litvinov IV, De Marzo AM, Isaacs JT
Nakayama M, De Marzo AM (2007) (also-keto reductase 1C2) and
(2003) Is the Achilles heel for
Abnormal DNA methylation, androgen metabolism in prostate
prostate cancer therapy a gain of
epigenetics, and prostate cancer. cells. Endocrinology 144:29222932
function in androgen receptor
signaling? J Clin Endorinol Metab Front Biosci 12:42544266 Scher HI, Beer TM, Higano CS, Anand
88:29722982 Normington K, Russell DW (1992) A, Taplin ME, Efstathiou E,
Tissue distribution and kinetic Rathkopf D, Shelkey J, Yu EY,
Litvinov IV, Vander Griend DJ,
characteristics of rat steroid 5a- Alumkal J, Hung D, Hirmand M,
Antony L, Dalrymple S, De Marzo
reductase isozymes. J Biol Chem Seely L, Morris MJ, Danila DC,
AM, Drake CG, Isaacs JT (2006)
267:1954819554 Humm J, Larson S, Fleisher M,
Androgen receptor as a licensing
Sawyers CL; Prostate Cancer
factor for DNA replication in Olsson AY, Valtonen-Andre C, Lilja H, Foundation/Department of Defense
androgen-sensitive prostate cancer Lundwall A (2004) The evolution of Prostate Cancer Clinical Trials
cells. Proc Natl Acad Sci USA the glandular kallikrein locus: Consortium (2010) Antitumour
103:1508515090 identification of orthologs and activity of MDV3100 in
Litvinov IV, Vander Griend DJ, Xu Y, pseudogenes in the cotton-top castration-resistant prostate cancer:
Antony L, Dalrymple SL, Isaacs JT tamarin. Gene 343:347355 a phase 12 study. Lancet
(2009) Low-calcium serum-free Page ST, Lin DW, Mostaghel EA, 375:14371446
medium selects for growth of Marck BT, Wright JL, Wu J, Amory Shah RB, Mehra R, Chinnaiyan AM,
normal prostate stem cells. Cancer JK, Nelson PS, Matsumoto AM Shen R, Ghosh D, Zhou M,
Res 66:85988607 (2011) Dihydrotestosterone Macvicar GR, Varambally S,
Mann T, Mann CL (1981) Male administration does not increase Harwood J, Bismar TA, Kim R,
Reproductive Function and Semen. intraprostatic androgen Rubin MA, Pienta KJ (2004)
Springer-Verlag, New York concentration or alter prostate Androgen-independent prostate
androgen action in heathy men: a cancer is a heterogeneous group of
Marx FJ, Karenberg A (2009) History
randomized-controlled trial. J Clin diseases: lessons from a rapid
of the term prostate. Prostate
Endrocrinol Metab 96:430437 autopsy program. Cancer Res
69:208213
Peters CA, Walsh PC (1987) The effect 64:92099216
McNeal JE, Bostwick DG (1986)
of nafaralin acetate, a luteinizing Silver SJ, Balmacoda J, Borrero C
Intraductal dysplasia: a
hormone-releasing hormone (1988) Pregnancy with sperm
premalignant lesion of the prostate.
agonist, on benign prostatic
Hum Pathol 17:6471 aspiration from the proximal head
hyperplasia. N Engl J Med of the epididymis: a new treatment
McNeal JE, Redwine EA, Freiha FS, 317:599604 for congenital absence of the vas
Stamey TA (1988) Zonal deferens. Fertil Steril 50:525530
Plymate SR, Tenover JS, Brenner WJ
distribution of prostatic
(1989) Circadian variation in Taplin ME, Bubley GJ, Ko TJ, Small EJ,
adenocarcinoma. Am J Surg Pathol
testosterone sex hormone-binding Upton M, Rajeshjuma B, Balk SP
12:897
globulin and calculated non-sex (1999) Selection for androgen
Meeker AK, Hicks JL, Platz EA, March hormone-binging globulin bound receptor mutations in prostate
GE, Bennett CJ, Delannoy MJ, De testosterone in healthy young and cancers treated with androgen
Marzo AM (2002) Telomere elderly man. J Androl 10:366371 antagonist. Cancer Res
shortening is an early somatic DNA 59:25112515
Price D, Williams-Ashman GH (1961)
alteration in human prostate
The accessory reproductive glands Taplin ME, Rajeshkumar B, Halabi S,
tumorigenesis. Cancer Res
of mammals. In: Young WC (ed) Werner CP, Woda BA, Picus J,
62:64066409
Sex and Internal Secretions, 3rd edn. Stadler W, Hayes DF, Kantoff PW,
Moore RA (1944) Benign hypertrophy Williams & Wilkins, Baltimore, Vogelzang NJ, Small EJ; Cancer and
and carcinoma of the prostate: MD, pp 366488 Leukemia Group B Study 9663

290
Chapter 13: Testosterone and the prostate

(2003) Androgen receptor human and mouse 17b- Verhagen AP, Ramaekers FC, Aalders
mutations in androgen-independent hydroxysteroid dehydrogenase type TW, Schaafsma HE, Debruyne
prostate cancer: cancer and 7. Biochem Biophys Res Commun FM, Schalken JA (1992)
Leukemia Group B Study 9663. 305:3745 Colocalization of basal and luminal
J Clin Oncol 21:26732678 cell-type cytokertins in human
Uemura M, Tamura K, Chung S,
Taylor BS, Schultz N, Hieronymus H, prostate cancer. Cancer Res
Honma S, Okuyama A, Nakamura
Gopalan A, Xiao Y, Carver BS, 52:61826187
Y, Nakagawa H (2008) Novel
Arora VK, Kaushik P, Cerami E, 5a-steroid reductase (SRD5A3, Wang Q, Carroll JS, Brown M (2005)
Reva B, Antipin Y, Mitsiades N, type-3) is overexpressed in Spatial and temporal recruitment
Landers T, Dolgalev I, Major JE, hormone-refractory prostate cancer. of androgen receptor and its
Wilson M, Socci ND, Lash AE, Cancer Sci 99:8186 coactivators involves chromosomal
Heguy A, Eastham JA, Scher HI, looping and polymerase tracking.
Vander Griend DJ, Litvinov IV, Isaacs
Reuter VE, Scardino PT, Sander C, Mol Cell 19:631642
Sawyers CL, Gerald WL (2010) JT (2007) Stabilizing androgen
Integrative genomic profiling of receptor in mitosis inhibits prostate Weihua Z, Makela S, Andersson LC,
human prostate cancer. Cancer Cell cancer proliferation. Cell Cycle Salmi S, Saji S, Webster JI, Jenson
18:1122 6:647651 EV, Nilsson S, Warner M,
Gustafsson J-A (2001) A role for
Thigpen AE, Davis DL, Milatovich A, Vander Griend DJ, Karthaus WL,
estrogen receptor beta in the
Mendonca BD, Imperato-McGinley Dalrymple S, Meeker A, DeMarzo
regulation of growth of the ventral
J, Griggin J, Franke U, Wilson JD, AM, Isaacs JT (2008) The role
prostate. Proc Natl Acad Sci USA
Russel DW (1992) Molecular of CD133 in normal human
98:63306335
genetics of steroid 5a-reductase 2 prostate stem cells and malignant
deficiency. J Clin Invest 90:799809 cancer-initiating cells. Cancer Res Weihua Z, Lathe R, Warner M,
68:97039711 Gustafsson J-A (2002) An endocrine
Thompson IM, Goodman PJ, Tangen pathway in the prostate ERb, AR,
CM, Lucia MS, Miller GJ, Ford LG, Vander Griend DJ, DAntonio J, Gurel
5a-androstane-3b ,17b-diol, and
Lieber MM, Cespedes RD, Atkins B, Antony L, DeMarzo AM, Isaacs
CYP7B1, regulates prostate growth.
JN, Lippman SM, Carlin SM, Ryan JT (2010) Cell-autonomous
Proc Natl Acad Sci USA
A, Szczepanek CM, Crowley JJ, intracellular androgen receptor
99:1358913594
Coltman CA Jr (2003) The signaling drives the growth of
influence of finasteride on the human prostate cancer initiating Wilson JD (1984) The endocrine
development of prostate cancer. cells. Prostate 70:9099 control of sexual differentiation.
N Engl J Med 349:215224 Harvey Lect 79:145172
van Leenders G, van Balken B, Aalders
Torn S, Nokelainen P, Kurlela R, T, Hulsbergen-van de Kaa C, Ruiter Xu Y, Dalrymple SL, Becker RE,
Pulkka A, Menjivar M, Ghosh S, D, Schalken J (2002) Intermediate Denmeade SR, Isaacs JT (2006)
Coca-Prados M, Peltoketo H, cells in normal and malignant Pharmacologic basis for the
Isomaa V, Vihko P (2003) prostate epithelium express c-MET: enhanced efficacy of dutasteride
Production purification and implications for prostate against prostatic cancers.
functional analysis of recombinant cancer invasion. Prostate 51:98107 Clin Cancer Res 12:40724079

291
Chapter
Clinical use of testosterone in hypogonadism

14 and other conditions


Eberhard Nieschlag and Hermann M. Behre

14.1 Use of testosterone in male 14.1.3.9 Erythropoiesis 300


hypogonadism 292 14.1.3.10 Liver function 301
14.1.1 Classification and symptoms 14.1.3.11 Lipid metabolism 301
of hypogonadism 294 14.1.3.12 Prostate 301
14.1.2 Initiation of substitution therapy 14.1.3.13 Bone mass 301
and choice of preparation 294 14.2 Treatment of delayed puberty
14.1.3 Surveillance of testosterone substitution in boys 302
therapy 297 14.3 Overall stature 302
14.1.3.1 Behavior and mood 298 14.4 Micropenis and microphallus 303
14.1.3.2 Sexuality 298 14.5 Ineffective use of testosterone in male
14.1.3.3 Phenotype 298 infertility 303
14.1.3.4 Blood pressure and cardiac 14.6 Contraindications to testosterone
function 299 treatment 303
14.1.3.5 Serum testosterone 299 14.7 Overall effect of testosterone 304
14.1.3.6 Serum dihydrotestosterone 300 14.8 Key messages 304
14.1.3.7 Serum estradiol 300
14.9 References 304
14.1.3.8 Gonadotropins 300

14.1 Use of testosterone in male chapters (10 and 11) are therefore dedicated to these
exciting developments.
hypogonadism The clinical signs and symptoms of all syndromes
The primary clinical use of testosterone is substitution and disease entities are predominantly due to a lack of
therapy for male hypogonadism. Hypogonadism may testosterone or its action. The most frequent disorders
be caused by lesions of the hypothalamo-pituitary requiring testosterone substitution are Klinefelter
system (secondary hypogonadism), the testes them- syndrome (incidence 1 in 500 men), Kallmann
selves (primary hypogonadism) or a mixture of both syndrome, isolated hypogonadotropic hypogonadism
as in LOH. Lesions in the target organs may also cause (IHH), late-onset hypogonadism (LOH), anorchia and
hypogonadism (see Chapters 3 and 19). An overview of pituitary insufficiency. Some disorders such as varico-
the various disease entities and syndromes is provided cele, orchitis, maldescended testes and Sertoli-cell-only
in Table 14.1, and for a detailed description the reader is syndrome may not, or only eventually, require testoster-
referred to the textbook in andrology by Nieschlag et al. one substitution. Although discrete endocrine alter-
(2010). In recent years it became clear that there are ations may be noted by laboratory tests in these
strong interrelationships between hypogonadism on the patients, the endocrine capacity of the Leydig cells
one side and the metabolic syndrome as well as cardio- remains high enough to maintain serum testosterone
vascular disorders on the other side; two special in the lower physiological range.

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

292
Chapter 14: Clinical use of testosterone

Table 14.1 Overview of disorders with male hypogonadism classified according to localization of cause

Hypothalamic-pituitary origin (hypogonadotropic syndromes = secondary hypogonadism)


Isolated hypogonadotropic hypogonadism (IHH) including Kallmann syndrome
Congenital adrenal hypoplasia
PraderLabhartWilli syndrome
LaurenceMoonBiedl syndrome
Constitutional delay of puberty
Pituitary insufficiency/adenomas
Pasqualini syndrome
Isolated lack of FSH
Biological inactive LH or FSH
Hyperprolactinemia
Hemochromatosis
Testicular origin (hypergonadotropic syndromes = primary hypogonadism)
Congenital anorchia
Acquired anorchia
Maldescended testes
Klinefelter syndrome
XYY syndrome
XX male
Gonadal dysgenesis
Testicular tumors including Leydig cell tumors
Varicocele
Sertoli-cell-only syndrome
General disease e.g. renal failure, liver cirrhosis, metabolic syndrome, diabetes, myotonia dystrophica
Disorders of sexual differentiation due to enzyme defects in testosterone biosynthesis or LH-receptor defects
(Leydig cell aplasia)
Exogenous factors
Mixed primary and secondary hypogonadism
Late-onset hypogonadism
Target organ resistance to sex steroids
Complete androgen insensitivity (CAIS) (testicular feminization)
Reifenstein syndrome (partial androgen insensitivity; PAIS)
Perineoscrotal hypospadias with pseudovagina
Aromatase deficiency
Estrogen resistance
Gynecomastia

293
Chapter 14: Clinical use of testosterone

In order to achieve fertility in patients with differentiation, leads to the development of intersexual
hypothalamic (IHH) or pituitary insufficiency, treatment genitalia (see Chapter 3). Lack of testosterone at the end of
with gonadotropins (hCG/hMG) or pulsatile GnRH is fetal life results in maldescended testes and small penis
required temporarily (e.g. Bchter et al. 1998; Depen- size. In later life the onset of testosterone deficiency before
busch et al. 2002; Warne et al. 2009). Once a pregnancy or after completion of puberty determines clinical appear-
has been induced these patients will go back on testoster- ance (Table 14.2).
one substitution. Individuals with hypogonadism of tes- If testosterone is lacking from the time of normal
ticular origin in whom infertility cannot be treated require onset of puberty onwards, eunuchoidal body proportions
testosterone substitution continuously. In all these will develop; i.e. arm span exceeds the standing height,
patients testosterone substitution is a lifelong therapy. and lower length of body (from soles to symphysis)
There is general agreement that patients with clas- exceeds upper length (from symphysis to top of the
sical disorders of primary or secondary hypogonadism cranium), and bone mass will not develop to its normal
should receive testosterone substitution therapy. How- level. The distribution of fat will remain prepubertal and
ever, there is a relatively large group of patients in whom feminine; i.e. emphasis of hips, buttocks and lower belly.
hypogonadism develops as a corollary of other acute or Voice mutation will not occur. The frontal hairline will
chronic diseases. Although these patients lack testoster- remain straight without lateral recession; beard growth is
one and show symptoms of hypogonadism, testosterone absent or scanty; the pubic hairline remains straight.
is usually not administered to them. Just why substitution Hemoglobin and erythrocytes will be in the lower-normal
is withheld is not quite clear. Probably in many phys- to subnormal range. Early development of fine perioral
icians minds testosterone is still predominantly associ- and periorbital wrinkles is characteristic. Muscles remain
ated with sexual functions. However, the better the underdeveloped. The skin is dry due to lack of sebum
general effects of testosterone on well-being, mood, production, and free of acne. The penis remains small; the
bones, muscles and red blood are understood, the more prostate is underdeveloped. Spermatogenesis will not be
frequently testosterone substitution will be considered. initiated, and the testes remain small. If an ejaculate can be
Chapter 17 is dedicated to the possible use of testosterone produced it will have a very small volume. Libido and
in these non-gonadal diseases. Similarly, LOH occurring normal erectile function will not develop.
with increasing incidence in aging men, and representing A lack of testosterone occurring in adulthood cannot
a combined form of primary and secondary hypogonad- change body proportions, but will result in decreased
ism, is associated with symptoms of testosterone defi- bone mass and osteoporosis, as well as in accumulation
ciency. But there is no general agreement on treatment of abdominal fat. Early-on, lower backache and, at an
strategies of this condition, and Chapter 16 deals with advanced stage, vertebral fractures may occur.
LOH and the controversies and unresolved problems Once mutation has taken place the voice will not
surrounding this area. Chapter 18 analyses and compares change again. Lateral hair recession and baldness when
the various guidelines for treatment of testosterone defi- present will persist, the secondary sexual hair will become
ciency issued by different societies and organizations. As scanty and, in advanced cases, a female hair pattern may
is stated there, most of the recommendations are not again develop. Mild anemia may develop. Muscle mass
strictly evidence based, and clinical experience prevails and power decrease. The skin will become atrophied and
as the major criterion. Therefore, for the time being, the wrinkled. Gynecomastia may develop. The prostate will
principle may be followed that any type of hypogonadism decrease in volume while the penis will not, or only
documented by decreased serum testosterone concentra- minimally, change its size, but will lose its function for
tions deserves testosterone substitution, unless there is a coitus (Chapter 12). Spermatogenesis will decrease and,
clear contraindication, of which there are only a few. as a consequence, also the size of the testes, which will
become softer. Libido and sexual arousability will
14.1.1 Classification and symptoms decrease or disappear, while potency will be less affected.

of hypogonadism 14.1.2 Initiation of substitution therapy


The time of onset of testosterone deficiency is of greater
importance for the clinical symptoms than localization of and choice of preparation
the cause. Lack of testosterone or testosterone action Testosterone substitution is started when the diagno-
during weeks 8 to 14 of fetal life, the period of sexual sis is established and serum testosterone levels below

294
Chapter 14: Clinical use of testosterone

Table 14.2 Symptoms of hypogonadism relative to age of manifestation

Affected organ/function Onset of lack of testosterone

Before After

completed puberty
Larynx No voice mutation No change
Hair Horizontal pubic hairline, straight frontal Diminishing secondary body hair,
hairline, diminished beard growth decreased beard growth
Skin Absent sebum production, lack of acne, Decreased sebum production, lack of acne,
pallor, skin wrinkling pallor, skin wrinkling, hot flashes
Bones Eunuchoid tall stature, arm span > height, Arm span = height, osteoporosis
osteoporosis
Bone marrow Low degree anemia Low degree anemia
Muscles Underdeveloped Atrophy
Prostate Underdeveloped Atrophy
Penis Infantile No change of size, loss of function
Testes Small volume, often maldescended testes Decrease of volume and consistency
Spermatogenesis Not initiated Arrest
Ejaculate Not produced Low volume
Libido Not developed Loss
Erectile function Not developed Erectile dysfunction

the normal range are found, taking into account the preparations have the advantage that at least some
various influences on serum testosterone levels, mimic the normal physiological diurnal rhythm and
including diurnal variations. In order to establish a thus represent the most physiological form of
diagnosis by documenting low serum testosterone substitution.
levels, usually determination of testosterone in a For full im substitution, pharmacokinetic and
serum sample taken between 07.00 and 11.00 in the clinical studies show that 200250 mg testosterone
morning is sufficient (Vermeulen and Verdonck enanthate or testosterone cypionate must be injected
1992). Pooled sera will not improve diagnostic accur- every two weeks (Nieschlag et al. 1976; Schulte-
acy (see Chapter 4). Beerbhl and Nieschlag 1980; Snyder and Lawrence
The symptoms of androgen deficiency can be pre- 1980; Sokol et al. 1982; Cunningham et al. 1990).
vented or reversed by testosterone treatment. It is More recently, testosterone undecanoate dissolved in
important that a preparation with natural testoster- castor oil and injected intramuscularly has been
one is selected for treatment so that all functions of shown to be effective in substitution therapy (Behre
testosterone and its active metabolites, DHT and et al. 1999a; von Eckardstein and Nieschlag 2002;
estradiol, can be exerted (Fig. 14.1). Of all testosterone Zitzmann and Nieschlag 2007; Brabrand et al. 2011).
preparations and routes of application described in Peak values remain within the normal range. In order
Chapter 15, im injection or oral ingestion of testoster- to achieve a steady state at the beginning of substitu-
one esters were formerly the most widely accepted tion, the second 1000 mg injection is given 6 weeks
and practiced modalities for the treatment of all after the first; further injections follow 1014 weeks
forms of hypogonadism. Over the last two decades, later. Individual intervals are determined according to
transdermal testosterone preparations have become a serum testosterone levels which are measured imme-
valuable alternative, first transdermal patches and, diately before the next injection. These determin-
more recently, transdermal gels. The transdermal ations are then repeated in yearly intervals. Values

295
Chapter 14: Clinical use of testosterone

Fig. 14.1 Threshold levels of serum


testosterone for various symptoms of
Total testosterone (nmol/l) late-onset hypogonadism (LOH) in 434
patients (Zitzmann et al. 2006).
20 See plate section for color version.

74
Normal

69
15

Loss of libido
84
Loss of vigor
12
Obesity 65
10
Feeling depressed, disturbed sleep
Lacking concentration 67
Diabetes mellitus type 2 (also non-obese men)
8
Hot flushes
Erectile dysfunction 75

that are too high lead to extension of injection inter- is not negatively affected, as was shown in 35 men
vals, and those that are too low to a shortening in taking 80200 mg testosterone undecanoate over 10
injection intervals. Slow intergluteal injections are years (Gooren 1994). The patients need to be
recommended. No adverse side-effects have been instructed to ingest the capsules together with a meal
observed, even after many years of use (Zitzmann in order to guarantee adequate absorption from the
and Nieschlag 2007). If the dose of testosterone unde- gut (Bagchus et al. 2003).
canoate injected is lowered, e.g. to 750 mg, the injec- Transdermal testosterone preparations mimic
tion intervals need to be shortened to maintain physiological diurnal variations, and their kinetic pro-
normal serum levels (Wang et al. 2010). file is closest to the ideal substitution. They may be
Testosterone pellet implants were among the first used as first choice and are especially well suited for
modalities applied for TRT (see Chapter 1). If three to patients who suffer from fluctuating symptoms
six implants are inserted, slowly declining serum tes- caused by other preparations. In addition, upon
tosterone levels in the normal range are achieved for removal, testosterone is immediately eliminated and
four to six months. There is, however, an initial burst they are therefore specifically suited for substitution
release, so that supraphysiological levels of about in advanced age (Wang et al. 2008).
50 nmol/l result. Commercially, pellets are only avail- Scrotal patches consisting of a thin film contain-
able in a few countries. ing 15 mg native testosterone were the first on the
If oral substitution is preferred, 40 mg testoster- market. They were applied daily in the evening and
one undecanoate capsules must be given two to four led to sufficient serum testosterone levels for 2224
times daily. These doses have been shown to be effect- hours. Adequate long-term substitution effect was
ive in the majority of hypogonadal men in either open achieved without serious side-effects under regular
(Franchi et al. 1978; Morales et al. 1997) or double- use, as has been observed in patients treated for up
blind controlled studies (Luisi and Franchi 1980; to 10 years with these patches (Behre et al. 1999b).
Skakkebaek et al. 1981) when libido and potency as Later developments superseded this initially useful
well as physical and mental activity were taken as preparation.
parameters. Although relatively high testosterone Non-scrotal transdermal systems also result in
doses are consumed with this regimen, liver function physiological serum levels with an appropriate

296
Chapter 14: Clinical use of testosterone

number of systems, which have to be applied in the Testosterone therapy does not prevent the chance of
evening. As resorption of testosterone depends on the initiating or reinitiating spermatogenesis with releas-
use of enhancers, in some cases considerable skin ing or gonadotropic hormones. Once spermatogen-
reactions limit the use of the systems. Although the esis has been induced it can be maintained for some
patches mentioned above are hardly used today, time with hCG alone, keeping intratesticular testos-
recently a new testosterone patch was developed caus- terone concentrations high (Depenbusch et al. 2002).
ing little skin irritation and which must be changed Patients with residual testosterone production
only every other day; however, two systems with may not require a full maintenance dose, e.g. Kline-
either 1.8 or 2.4 mg resorbed per day must be used felter patients in an early phase of testosterone
(Raynaud et al. 2009). deficiency. In these cases injection intervals of testos-
A further transdermal application is the use of terone esters may be extended; these cases may also be
testosterone gels, which are applied to large skin areas suited for low-dose testosterone undecanoate therapy
in order to allow sufficient amounts of the hormone (i.e. 40 mg once or twice daily) or intermittent trans-
to be resorbed. These gels are applied in the morning dermal treatment (e.g. every second or third day).
to the upper arm, shoulders and abdomen and are left This dose does not entirely suppress the residual
to dry for five minutes. During this time contact with endogenous testosterone production and supplements
women or children, direct as well as through wash- the lacking hormone.
rags and towels (de Ronde 2009), must be avoided, Finally, the question needs to be addressed at
because of the danger of contamination. Thereafter which serum testosterone levels substitution therapy
the danger is negligible especially if the skin is washed should be initiated. Based on clinical experience and
after evaporation of the alcohol. Physiological levels many studies in normal and hypogonadal men, we
result when the gel is applied in the morning. Long- considered 1240 nmol/l as the normal range for
term use over several years showed good results many years (Nieschlag et al. 2010), i.e. starting treat-
(Wang et al. 2004; McNicholas and Ong 2006). If a ment when in the presence of relevant symptoms
preparation with a higher testosterone concentration testosterone levels dropped below 12 nmol/l. How-
is used, less gel needs to be applied and good clinical ever, in other countries different levels were con-
results are obtained. If this gel is applied to the scro- sidered to be the limit to start substitution; e.g. 7.5
tum, only one fifth of the amount required when used nmol/l in France, 7.5 to 8.0 nmol/l in the UK and
on other skin areas is necessary for substitution (Kh- 9.0 nmol/l in Spain (Nieschlag et al. 2004). This
nert et al. 2005). However, the scrotal application has prompted a systematic investigation in patients sus-
not yet been licensed. pected of LOH, and symptom-specific testosterone
The choice of testosterone preparation and route threshold levels resulted (Fig. 14.1). With regard to
of administration is ultimately up to the patient, who the different lower limits of normal, this implies that
over time may gather experience with several prepar- physicians consider different symptoms as an indica-
ations and develop his own preference. Younger tion to start substitution, and the lower limit of
patients will be more inclined to choose long-acting 12 nmol/l could be raised rather than lowered. The
preparations, while the older patient (> 50 years) 12 nmol/l threshold received further support from a
should be advised to use a short-acting preparation recent study on reference ranges for normal men
at least initially (Wang et al. 2008). If therapy has to based on several cohorts and liquid chromatog-
be stopped due to developing contraindications (e.g. raphytandem mass spectrometry leading to
prostate disease), serum testosterone levels will imme- 12.1 nmol/l (= 348 ng/dl) as the lower limit of normal
diately decline to endogenous levels. (2.5th percentile (Bhasin et al. 2011)). In the same
If a patient has pronounced androgen deficiency, study, 243 pmol/l (= 70 pg/dl) was estimated to be the
has never received testosterone and has passed the age lower limit of free testosterone.
of puberty, he is immediately treated with a full
maintenance dose of testosterone. In cases of second- 14.1.3 Surveillance of testosterone
ary hypogonadism when fertility is requested, testos-
terone therapy can be interrupted and GnRH or hCG/ substitution therapy
hMG therapy can be implemented until sperm The physiological effects of testosterone can be used
counts increase and a pregnancy has been induced. for monitoring the efficacy of testosterone

297
Chapter 14: Clinical use of testosterone

substitution therapy (Table 14.2). Since therapy aims testosterone preparations avoid supraphysiological
at replacing the testosterone endogenously lacking, testosterone serum levels. Decreasing the testosterone
and since physiological serum concentrations are well dose is the rational consequence, but intervention by
known, serum testosterone levels also provide a good aspirating blood from the corpora cavernosa may be
parameter for therapy surveillance. Guidelines for acutely necessary.
monitoring testosterone therapy in general were first
issued by the World Health Organization (1992), 14.1.3.3 Phenotype
followed by various societies and organizations as Muscles and physical strength grow under testoster-
summarized in Chapter 18. one treatment, and the patient develops a more vigor-
ous appearance (e.g. Wittert et al. 2003). The increase
14.1.3.1 Behavior and mood in lean body mass at the expense of body fat usually
The patients general well-being is a good parameter results in a decrease of body weight (Rolf et al. 2002).
to monitor the effectiveness of replacement therapy. The distribution of subcutaneous fat that shows
Under sufficient testosterone replacement the patient feminine characteristics in hypogonadism (hips,
feels physically and mentally active, vigorous, alert lower abdomen, nates) may change with increasing
and in good spirits; too low testosterone levels will muscle mass. In particular, testosterone reduces
be accompanied by lethargy, inactivity and depressed abdominal fat.
mood (Burris et al. 1992; Wang et al. 1996; Zitzmann The appearance and maintenance of a male sexual
and Nieschlag 2001) (see also Chapter 5). hair pattern is a good parameter for monitoring tes-
tosterone replacement (see Chapter 7). In particular,
14.1.3.2 Sexuality beard growth and frequency of shaving can easily
The presence and frequency of sexual thoughts and be recorded. Hair growth in the upper pubic triangle
fantasies correlate with appropriate testosterone sub- is an important indicator of sufficient androgen
stitution; while loss of libido and sexual desire are a substitution. While women, boys and untreated
sign of subnormal testosterone values. Spontaneous hypogonadal patients have a straight frontal hairline,
erections such as those during the morning phase of androgenization is accompanied by temporal reces-
sleep will not occur if testosterone replacement is sion of the hairline and if a predisposition exists
inadequate; however, erections due to tactile or visual by the development of baldness. The pattern of male
erotic stimuli may be present even with low testoster- sexual hair is of greater importance than the intensity
one levels. The frequency of ejaculations and sexual of hair growth. The AR polymorphism plays a role
intercourse correlate with serum testosterone levels in in male hair growth and pattern (see Chapter 3).
the normal to subnormal range. Therefore, detailed A well-substituted patient may have to shave daily.
psychological exploration or a diary on sexual activity However, if there is no genetic disposition for dense
and interests are useful adjuncts in assessing testoster- beard growth, additional testosterone will not
one substitution. For objective evaluation of psycho- increase facial hair.
sexual effects, weekly questionnaires on sexual Sebum production correlates with circulating tes-
thoughts and fantasies, sexual interest and desire, tosterone levels, and hypogonadal men may suffer
satisfaction with sexuality, frequency of erections from dry skin. In an early phase of treatment patients
and number of morning erections and ejaculations may even complain about the necessity of shampoo-
may be used (e.g. Lee et al. 2003; Rosen et al. 2011). ing more frequently; they have to be informed that
These questionnaires are specifically suited for moni- this is a part of normal maleness. The occurrence of
toring substitution therapy and less for diagnosing acne may be a sign of supraphysiological testosterone
hypogonadism. levels, and the dose should be reduced accordingly.
Priapism has been reported to occur in individual Gynecomastia may be caused by increased con-
cases, mostly at the beginning of testosterone substi- version to estradiol during testosterone therapy, espe-
tution in adult patients as well as in boys with delayed cially under testosterone enanthate injections. After
puberty (Endres et al. 1987; Zelissen and Stricker initiation of androgen therapy and consequent
1988; Ruch and Jenny 1989; Arrigo et al. 2005; decrease of estradiol serum levels, gynecomastia usu-
Ichioka et al. 2006). This is an extremely rare effect ally disappears. If gynecomastia pre-exists due to an
and may have become even rarer since the new increased estradiol/testosterone ratio in hypogonadal

298
Chapter 14: Clinical use of testosterone

men, it may decrease during adequate testosterone such side-effects are not observed (e.g. Whitworth
therapy. However, in severe cases mastectomy by an et al. 1992). To the contrary, systolic and diastolic
experienced plastic surgeon may be required. blood pressure decrease under regular testosterone
Patients who have not undergone pubertal devel- substitution. Regular blood pressure measurement
opment will experience voice mutation soon after should be performed during testosterone therapy,
initiation of testosterone therapy (Akcam et al. especially during inception of treatment when the
2004). During normal pubertal development the voice testosterone dosages have to be adjusted, and in
begins to break when serum testosterone levels reach men with additional problems of the heart and
about 10 nmol/l and SHBG drops (Pedersen et al. kidneys. In patients with severe cardiovascular
1986). Mutation of the voice is very reassuring for problems testosterone must be administered very
the patient and helps him to adjust to his environ- carefully and in low doses (Basaria et al. 2010;
ment by closing the gap between his chronological Aaronson et al. 2011; Chapter 11).
and biological age. It is specifically important for the
patient to be recognized as an adult male on the 14.1.3.5 Serum testosterone
phone. Once the voice has mutated it is no longer a When serum testosterone levels are used to judge the
useful parameter for monitoring replacement therapy quality of testosterone substitution it is necessary to
since the size of the larynx, the vocal chords and thus be aware of the pharmacokinetic profiles of the dif-
the voice achieved will be maintained without requir- ferent testosterone preparations (Chapter 15). More-
ing further androgens. over, in longitudinal surveillance of testosterone
In patients who have not gone through puberty, therapy it is important to use assay systems that
penis growth will be induced by testosterone treat- strictly undergo internal and external quality control
ment and normal erectile function will develop. Since (Chapter 4). Generally, testosterone serum levels
penile androgen receptors diminish during puberty, should be measured just before the injection of the
growth will cease even under continued testosterone next dose of long-acting preparations or transdermal
treatment (Shabsigh 1997; see Chapter 12). application. The time point of the last injection or
Patients who did not undergo puberty before the administration of oral or transdermal testosterone
onset of hypogonadism may also develop eunuchoidal must be recorded to interpret the serum levels
body proportions because of retarded closure of the measured.
epiphyseal lines of the extremities. Testosterone treat- Levels below the lower normal limit at the end of a
ment will briefly stimulate growth, but will then lead three-week interval after testosterone enanthate injec-
to closure of the epiphyses and will arrest growth. In tion should prompt shorter injection frequency of
these patients, an X-ray of the left hand and distal end two-week intervals. Conversely, if the levels are in
of the lower arm should be made before treatment to the high physiological range at the end of the injec-
determine bone age. The epiphyseal closure may be tion interval, the dosing intervals may be extended.
followed by further X-rays during the course of treat- When using the im depot preparation of testoster-
ment. In addition, body height and arm span as one undecanoate, peak values remain within the
measured from the tip of the right to the tip of the left normal range. In order to achieve a steady state at
middle finger should be measured until no further the beginning of substitution the second 1000 mg
growth occurs. Continued growth, in particular of the injection is given 6 weeks after the first; further injec-
arm span, indicates inadequate androgen substitution tions follow 10 to 14 weeks later. Individual intervals
or extremely rare cases of estrogen resistance or aro- are determined according to serum testosterone
matase deficiency (see Chapters 8 and 19). levels, which are measured immediately before the
next injection. These determinations are then
14.1.3.4 Blood pressure and cardiac function repeated in yearly intervals. Values that are too high
Overdosing androgens, as can be observed during should lead to extension of injection intervals, those
misuse of testosterone and anabolic steroids, may that are too low to a shortening of injection intervals
increase blood pressure by increasing blood electro- (Zitzmann and Nieschlag 2007).
lytes and water retention, leading in extreme cases Low serum testosterone levels two to four hours
to edema (see Chapter 25). During effective testos- after ingestion of oral testosterone undecanoate
terone substitution therapy in hypogonadal men should prompt counseling of the patient so that the

299
Chapter 14: Clinical use of testosterone

capsule is taken together with a meal and testosterone 14.1.3.8 Gonadotropins


is better absorbed. However, it is difficult to base The determination of LH and FSH plays a key role
monitoring of treatment with oral testosterone unde- in establishing the diagnosis of hypogonadotropic
canoate on serum testosterone levels, and other par- (i.e. secondary) or hypergonadotropic (i.e. primary)
ameters are of more importance if this mode of hypogonadism. However, during surveillance of testos-
therapy is chosen. terone therapy they are of less importance. Negative-
When transdermal preparations are applied, feedback regulation between hypothalamus, pituitary
serum testosterone levels may be measured just before and testes causes negative correlation between serum
the next dose is administered. Initial measurements, testosterone and LH, as well as to some extent to FSH
however, are only meaningful after two or three weeks levels in normal men.
following initiation of therapy, since it takes time In cases with primary hypogonadism (e.g. intact
until the skin builds up a reservoir and steady state hypothalamic and pituitary function), FSH and in
serum levels are reached. particular LH increase with decreasing testosterone
After initiation of testosterone substitution, meas- levels and may normalize under testosterone treat-
uring serum testosterone under the conditions men- ment. This is especially the case in patients with
tioned above is recommendable after 3 to 6 and 12 acquired anorchia (e.g. due to accidents or iatrogenic
months, and thereafter annually. castration). However, in the most frequent form of
In blood, testosterone is bound to SHBG and primary hypogonadism, i.e. in patients with Klinefelter
other proteins. Only about 2% of testosterone is syndrome, LH and FSH often do not show significant
not bound and is available for biological action of suppression during testosterone substitution. More-
testosterone (free testosterone). Since total testoster- over, oral or transdermal testosterone may have only
one correlates well with free testosterone, separate little effect on gonadotropins. Therefore LH is not a
determination of free testosterone is not necessary good indicator of sufficient TRT.
for routine monitoring (see also Chapter 4).

14.1.3.9 Erythropoiesis
14.1.3.6 Serum dihydrotestosterone
Since erythropoiesis is androgen dependent, hypogo-
Determination of DHT does not play a role in rou-
nadal patients usually present with mild anemia (with
tine monitoring of TRT, but may be of importance
values in the female normal range) which normalizes
in experimental use of testosterone preparations and
under testosterone treatment. Therefore, hemoglobin,
monitoring biological effects of androgens. Due to
red blood cell count and hematocrit are good param-
the high 5a-reductase activity in skin, transdermal
eters for surveillance of replacement therapy. If suffi-
testosterone application is associated with increased
cient stimulation is lacking despite adequate
serum DHT levels; this applies especially to scrotal
testosterone therapy, lack of iron should be ruled
application. The DHT adds to the overall androgeni-
out and treated if necessary. At the beginning of
city of the preparation, and a patient receiving trans-
therapy red blood values should be assessed every
dermal treatment may be well substituted clinically
three months, and later on annually. If too much
although his serum testosterone does not reflect this.
testosterone is administered, supraphysiological levels
In these cases occasional measurement of serum
of hemoglobin, erythrocytes and hematocrit as a sign
DHT may be indicated (Khnert et al. 2005; see also
of polycythemia can develop, indicating that the tes-
Chapter 4).
tosterone dose should be scaled down (Calof et al.
2005). In some cases phlebotomy may be required
14.1.3.7 Serum estradiol acutely. The erythropoietic response not only depends
In sensitive patients, very high serum testosterone on the serum testosterone levels, but also on the age of
levels, as may occur under testosterone enanthate, the patient and androgen receptor polymorphism.
may be converted to estrogens and cause gynecomas- Older patients and those with shorter CAG repeats
tia. This is an indication to reduce the dose or switch react more sensitively to testosterone (Zitzmann and
to another testosterone preparation. In this case Nieschlag 2007).
monitoring serum estradiol levels may explain the Testosterone has been claimed to potentiate sleep
clinical findings. apnea (see Chapter 17); however, only case reports

300
Chapter 14: Clinical use of testosterone

about the incidence of sleep apnea during testosterone 14.1.3.12 Prostate


treatment have been published and, paradoxically, Before initiating testosterone treatment a prostate
hypogonadism has also been cited as a cause of carcinoma must be excluded. This is done by digital
this condition (Luboshitzky et al. 2002; Attal and rectal examination (DRE) and PSA determination.
Chanson 2010). Increased hematocrit and increased Imaging techniques such as transrectal ultrasonogra-
mass of pharyngeal muscle bulk, as well as neuroen- phy (TRUS) are not considered mandatory, but may
docrine effects of testosterone during therapy were add information.
discussed as possible reasons. The development of Prostate volume as determined by transrectal
signs and symptoms of obstructive sleep apnea during ultrasonography is a sensitive end-organ parameter
testosterone therapy warrants a formal sleep study for surveillance. Testosterone substitution therapy
and treatment with continuous positive airway pres- increases prostate volume in hypogonadal men, but
sure (CPAP) if necessary. If the patient is unrespon- only to the extent seen in age-matched controls
sive or cannot tolerate continuous positive airway (Behre et al. 1994). Prostate volume growth also
pressure, the testosterone must be reduced or depends on the AR polymorphism (Zitzmann et al.
discontinued. 2003). Prostate-specific antigen increases slightly
during therapy but remains within the normal range
14.1.3.10 Liver function (Behre et al. 1999a; Zitzmann and Nieschlag 2007).
The testosterone preparations proposed for testoster- Since testosterone therapy must be terminated if a
one replacement do not have negative side-effects on prostate carcinoma occurs and prostate carcinoma is
liver function. Nevertheless, many physicians believe a disease of advanced age, patients above 45 years of
that testosterone may disturb liver function. This age under testosterone treatment should be regularly
impression derives from 17a-methyltestosterone and investigated, first after 36 months and 12 months
other 17a-alkylated anabolic steroids which are and then at yearly intervals (Wang et al. 2008). Meas-
indeed liver toxic and which should no longer be used urement of PSA and palpation of the prostate, if
in the clinic (see Chapter 25). possible supported by transrectal ultrasonography,
Monitoring liver function is of special interest in should be performed. As a sign of adequate prostate
hypogonadal patients with concomitant diseases that and seminal vesicle stimulation, ejaculate volume will
affect liver function, or in patients whose hypogonad- increase into the normal range.
ism is induced by general diseases. In such cases
additional medication is necessary that may influence
liver function and thus influence testosterone metab- 14.1.3.13 Bone mass
olism, e.g. by increasing SHBG production. Testosterone replacement therapy in hypogonadal
men will increase the low bone mineralization, pre-
14.1.3.11 Lipid metabolism venting or reversing osteoporosis and possibly bone
Lipid profiles may change under testosterone substi- fractures (Snyder 1999; Chapter 8). In particular,
tution. Presumed adverse effects such as decreasing with respect to bones it is important to use testoster-
high-density lipoprotein (HDL) levels and increasing one preparations that can be converted into estro-
low-density lipoprotein (LDL) levels have been gens since this hormone plays a significant role in
reported when comparing different treatment modal- bone metabolism (Ohlsson and Vandenput 2009).
ities (Jockenhvel 1999). However, beneficial effects Bone density should be measured in patients receiv-
were also seen, especially in older hypogonadal men, ing testosterone substitution, prior to treatment and
as LDL levels decreased under testosterone substitu- regularly every two years as long as treatment con-
tion and HDL increased. The CAG repeat length of tinues. Quantitative computer tomography (QCT) of
the androgen receptor has a modifying role in the the lumbar spine provides accurate information;
effects on lipid parameters (Zitzmann et al. 2003), other validated methods are dual photon absorpti-
and pharmacogenetic considerations may in the ometry and dual energy X-ray absorptiometry. Also
future influence dose and route of testosterone sonographic measurement of bone density (for
administration. Currently, it appears sufficient to example, of the phalangi) provides a useful and
monitor lipids under testosterone therapy in those inexpensive parameter for monitoring (Zitzmann
patients with grossly abnormal lipid profiles. et al. 2002).

301
Chapter 14: Clinical use of testosterone

14.2 Treatment of delayed puberty have testicular growth induced by androgen therapy.
Because pubertal growth is a product of the interaction
in boys of growth hormone (GH) and insulin-like growth
Androgen replacement therapy in male adolescents factor 1 (IGF-1) and the hypothalamic-pituitary-
with constitutional delay of growth and puberty has gonadal axis, boys with concomitant GH deficiency
been shown to be beneficial psychologically as well as will require the simultaneous administration of GH
physiologically, and should be initiated promptly on and androgens for the treatment of delayed puberty.
diagnosis (de Lange et al. 1979; Rosenfeld et al. 1982; In boys with secondary causes of delayed puberty,
Albanese and Stanhope 1995; Rogol 2005). Boys with development can also be induced by pulsatile GnRH
delayed puberty are at risk for not obtaining adequate or hCG/hMG respectively. This therapy has the
peak bone mass and for having deficiencies in advantage that testicular development is induced
developing social skills, an impaired body image and simultaneously. However, we prefer to induce initial
low self-esteem. Younger boys with short stature, virilization by testosterone and to stimulate sperm-
delayed bone age (at least 10.5 years), and delayed atogenesis at a later stage with the more demanding
pubertal development in the absence of other endo- GnRH or gonadotropin therapy.
crinological abnormalities can be treated with
50100 mg of testosterone enanthate or cypionate
im, every four weeks for three months, whereas 14.3 Overall stature
boys >14 years old may be treated with 250 mg (im, The effect of testosterone on epiphyseal closure may be
every four weeks for three months). After a three- used to treat boys who are dissatisfied with their pro-
month wait and see period, another course of treat- spective final overall body height (for review see Drop
ment may be offered if pubertal development does not et al. 1998). Treatment has to start before the age of 14.
continue. An increase in testes size is the most import- Doses of 500 mg testosterone enanthate have to be
ant indicator of spontaneous pubertal development administered every two weeks for at least a year to
(testes volume >3 ml). Overtreatment with testoster- produce effects (Bettendorf et al. 1997). This treatment
one may result in premature closure of the epiphyses of should be reserved for special cases since tall stature is
long bones, resulting in reduced adult height. There- not a disease but rather a cosmetic and psychological
fore, treatment of patients who have not yet reached full problem. However, social and psychological conflicts
adult height has to be undertaken carefully. caused by this condition should not be underesti-
Low-dose oral testosterone undecanoate has been mated. It should also be remembered that testosterone
tested for the treatment of constitutional delay of is not registered for this treatment, which has therefore
puberty (Albanese et al. 1994; Brown et al. 1995). to be considered experimental. Combining ethinyl
For example, treatment of 1114-year-old prepubertal estradiol with testosterone injections has no additional
boys with 20 mg testosterone undecanoate per day for height-reducing effect (Decker et al. 2002).
six months resulted in an increase in growth velocity An additional reservation comes from the possible
without advancing bone age and pubertal develop- effects of such high-dose testosterone treatment at
ment (Brown et al. 1995). Such mild treatment this early age on fertility, the prostate, the cardiovas-
appears to be suited for an early phase when viriliza- cular system, on bones and other organs. Long-term
tion is not yet requested. Transdermal testosterone follow-up of men treated on average 10 years earlier
should also be a useful method to induce puberty. with high-dose testosterone for tall stature revealed
However, experience in a larger series of patients has no negative effects on sperm parameters and repro-
not yet been reported. ductive hormones in comparison to controls (de Waal
At the beginning of therapy it is often difficult to et al. 1995; Lemcke et al. 1996; Hendriks et al. 2010).
distinguish between boys with constitutional delay of Prostate morphology as evaluated by ultrasonography
growth and puberty, who require only temporary did not show any abnormalities, and serum lipids
androgen replacement, and boys with idiopathic hypo- were not different from the control group. Slightly
gonadotropic hypogonadism, who require lifelong lower sperm motility was attributable to a higher
androgen therapy to stimulate puberty and to main- incidence of varicocele and maldescended testes in
tain adult sexual function. However, boys with per- the treated men rather than to the treatment as such.
manent hypogonadotropic hypogonadism will not Thus it appears that, as far as evaluated, high-dose

302
Chapter 14: Clinical use of testosterone

treatment has no long-term negative side-effects Thus, to date testosterone and other androgens
in these adolescents. have no place in evidence-based treatment of idio-
pathic male infertility (Kamischke and Nieschlag
1999).
14.4 Micropenis and microphallus
Enlargement of a micropenis or microphallus can be
achieved in children by treatment with 2550 mg of 14.6 Contraindications to
testosterone enanthate or cypionate (im, every three testosterone treatment
to four weeks for three months; Ishii et al. 2004) or Effects and side-effects of testosterone therapy have
with 1.255% testosterone cream, 5% DHT cream or been described in detail above. Here the major
10% testosterone propionate cream (twice daily for reasons for not initiating or for interrupting testoster-
three months). High-dose androgen therapy may be one therapy are briefly summarized.
necessary to achieve some androgenization in male The major contraindication to testosterone ther-
pseudohermaphroditism caused by 5a-reductase defi- apy is a prostate carcinoma. A patient with an existing
ciency and certain AR defects. prostate carcinoma should not receive testosterone.
A carcinoma has to be excluded before starting ther-
14.5 Ineffective use of testosterone apy, and the patient on testosterone should be
checked regularly for prostate cancer (digital explor-
in male infertility ation, PSA, transrectal sonography and biopsy, if
Since testosterone has been used so effectively in the necessary). (See also Chapter 13.)
treatment of endocrine insufficiency of the testes, its Breast cancer cells often are hormone sensitive,
use has also been attempted in the treatment of idio- especially estrogen sensitive, and therefore, for
pathic male infertility. Testosterone rebound was one reasons of safety, breast cancer is considered a con-
of the earliest modalities in this regard. The published traindication to testosterone treatment. However,
success rate in terms of pregnancies varied consider- breast cancer is a relatively rare cancer in men and
ably from center to center, but remained low overall no cases of testosterone substitution and occurrence
(Charny and Gordon 1978). All studies were uncon- of breast cancer have been published, as an extended
trolled trials without placebo and double-blinding, literature search revealed. Thus, this warning cannot
and therefore inconclusive. Testosterone rebound be substantiated.
therapy cannot be recommended for treatment of In some countries sexual offenders may be or have
infertility and is no longer practiced. been treated by castration or antiandrogenic therapy.
More recently, testosterone undecanoate has been It would be a serious mistake to administer testoster-
tested for the treatment of idiopathic male infertility. one to such patients. Relapses and renewed crimes
However, a significant increase in pregnancy rates could be the consequence and the responsibility of
could not be demonstrated (Pusch 1989; Comhaire the prescribing physician. The same holds true for
et al. 1995). When testosterone undecanoate was pharmacological androgen deprivation therapy
given combined with tamoxifen and/or hMG, an (ADT) using GnRH analogs or antiandrogens, which
improvement of semen parameters was observed is currently the preferred therapy for sexual offenders.
(Adamopoulos et al. 1995; 1997). However, in these Testosterone suppresses spermatogenesis: a phe-
studies no pregnancy rates were reported. The thera- nomenon exploited for hormonal male contraception
peutic goal of every infertility treatment should be an (see Chapter 23). In hypogonadal patients with
increase in pregnancy rates; therefore, studies in reduced spermatogenetic function, testosterone
which only improved semen parameters are administration will also decrease sperm production.
reported, without examining the pregnancy rates, Such patients who wish to father children, e.g. by
must be considered as inconclusive in terms of infer- techniques of artificial fertilization, should not receive
tility treatment. Similarly, after many years of clinical testosterone substitution therapy, at least not for the
use, no significant effect of mesterolone on preg- time their sperm are necessary for fertilization of
nancy rates could be demonstrated in an extensive eggs. This is of increasing importance as not only
WHO-sponsored multicenter trial (World Health residual sperm in patients with secondary hypogo-
Organization 1989). nadism but also with Klinefelter syndrome may be

303
Chapter 14: Clinical use of testosterone

able to fertilize eggs via intracytoplasmatic sperm


injection (ICSI) and induce pregnancies (e.g. hypogonadism. For substitution,
testosterone preparations should be used that
Lanfranco et al. 2004).
can be converted to 5a-dihydrotestosterone
(DHT) as well as to estradiol, in order to
14.7 Overall effect of testosterone develop the full spectrum of testosterone
action.
Testosterone has many biological functions and, as
 Injectable, oral and transdermal testosterone
demonstrated in this chapter, testosterone is a safe preparations are available for clinical use.
medication. There are only very few reasons why The best preparation is the one that
testosterone should be withheld from a hypogonadal replaces testosterone serum levels at as
patient (see Section 14.6). Nevertheless, to date many close to physiological concentrations as
hypogonadal men still do not receive the benefit of possible. This objective is best reached by
testosterone therapy because they are not properly testosterone gels and by injectable testosterone
diagnosed and the therapeutic consequences are not undecanoate.
drawn (e.g. Bojesen et al. 2003). Some physicians even  In seven decades of clinical use testosterone has
believe that the shorter life expectancy of men com- proven to be a very safe medication. No toxic
pared to women could be attributed to effects of effects are known. The only important
contraindication is the presence of a prostate
testosterone. However, large epidemiological studies
carcinoma which should be excluded before
of healthy men or of patients have demonstrated that substitution is initiated.
men with lower testosterone serum levels have a  Testosterone therapy should be monitored
shorter life expectancy than those with higher testos- by patients well-being, alertness and
terone levels (Shores et al. 2006; Laughlin et al. 2008; sexual activity, by occasional measurement
Haring et al. 2010; Bojesen et al. 2011). It remains of serum testosterone levels, hemoglobin and
currently unresolved whether testosterone levels are hematocrit, by bone density measurements
just an indicator of the general health status or and prostate parameters (rectal examination,
whether there is a causal positive relationship between PSA and transrectal sonography).
testosterone levels and longevity. By the same token it  Testosterone can be used to initiate puberty
remains unresolved today whether testosterone treat- in boys with constitutional delay of pubertal
development. Careful dosing does not lead to
ment of hypogonadal men may extend or shorten
premature closure of the epiphysis and reduced
their life expectancy. Without doubt, however, testos- height.
terone substitution significantly improves the quality  High-dose testosterone treatment in early
of life of hypogonadal men. puberty may prevent expected over-tall stature in
boys. Negative long-term effects of this
14.8 Key messages treatment have not become evident to date.
 The primary indications for testosterone  Testosterone treatment is not indicated in
therapy are the various forms of male idiopathic male infertility.

14.9 References tamoxifen citrate in idiopathic


oligozoospermia. Fertil Steril
Akcam T, Bolu E, Merati AL, Durmus C,
Gerek M, Ozkaptan Y (2004)
Aaronson AJ, Morrissey RP, Nguyen 64:818824 Voice changes after androgen therapy
CT, Willix R, Schwarz ER (2011) for hypogonadotrophic
Update on the safety of testosterone Adamopoulos DA, Nicopoulou St,
Kapolla N, Karamertzanis M, hypogonadism. Laryngoscope
therapy in cardiac disease. Expert 114:15871591
Opin Drug Saf 10:697704 Andreou E (1997) The
combination of testosterone Albanese A, Stanhope R (1995)
Adamopoulos DA, Nicopoulou S, undecanoate with tamoxifen Predictive factors in the
Kapolla N, Vassipoulos P, citrate enhances the effects of determination of final height in
Karamertzanis M, Kontogeorgos L each agent given independently boys with constitutional delay of
(1995) Endocrine effects of on seminal parameters in growth and puberty. J Pediatr
testosterone undecanoate as a men with idiopathic 126:545550
supplementary treatment to oligozoospermia. Fertil Steril
menopausal gonadotropins or Albanese A, Kewley GD, Long A,
67:756762 Pearl KN, Robins DG, Stanhope R

304
Chapter 14: Clinical use of testosterone

(1994) Oral treatment for Bettendorf M, Heinrich UE, Burris AS, Banks SM, Carter CS,
constitutional delay of growth and Schnberg DK, Grulich-Henn J Davidson JM, Sherins RJ (1992)
puberty in boys: a randomized trial (1997) Short-term, high dose A long-term prospective study of
of an anabolic steroid or testosterone treatment fails to the physiologic and behavioural
testosterone undecanoate. Arch Dis reduce adult height in boys with effects of hormone replacement in
Child 71:315317 constitutional tall stature. Eur untreated hypogondadal men.
Arrigo T, Crisafulli G, Salzano G, J Pediatr 156:911915 J Androl 14:297304
Zirilli G, De Luca F (2005) High- Bhasin S, Pencina M, Jasuja GK, Calof OM, Singh AB, Lee ML, Kenny AM,
flow priapism in testosterone- Travison TG, Coviello A, Orwoll E, Urban RJ, Tenover JL, Bhasin S (2005)
treated boys with constitutional Wang PY, Nielson C, Wu F, Tajar A, Adverse events associated with
delay of growth and puberty may Labrie F, Vesper H, Zhang A, Ulloor testosterone replacement in middle-
occur even when very low doses are J, Singh R, DAgostino R, Vasan RS aged and older men: a meta-analysis
used. J Endocrinol Invest (2011) Reference ranges for of randomized, placebo-controlled
28:390391 testosterone in men generated using trials. J Gerontol A Biol Sci Med Sci
Attal P, Chanson P (2010) Endocrine liquid chromatography tandem 60:14511457
aspects of obstructive sleep apnea. mass spectrometry in a community- Charny CW, Gordon JA (1978)
J Clin Endocrinol Metab 95:483495 based sample of healthy nonobese Testosterone rebound therapy:
young men in the Framingham heart a neglected modality. Fertil Steril
Bagchus WM, Hust R, Maris F, study and applied to three
Schnabel PG, Houwing NS (2003) 29:6468
geographically distinct cohorts.
Important effect of food on the J Clin Endocrinol Metab 96: Comhaire F, Schoonjans F,
bioavailability of oral testosterone 24302439 Abelmassih R, Gordts S, Campo R,
undecanoate. Pharmacotherapy Dhont M, Milingos S, Gerris J
23:319325 Bojesen A, Juul S, Gravholt CH (2003) (1995) Does treatment with
Prenatal and postnatal prevalence of testosterone undecanoate improve
Basaria S, Coviello AD, Travison TG, Klinefelter syndrome: a national
Storer TW, Farwell WR, Jette AM, the in-vitro fertilizing capacity of
registry study. J Clin Endocrinol spermatozoa in patients with
Eder R, Tennstedt S, Ulloor J, Zhang Metab 88:622626
A, Choong K, Lakshman KM, Mazer idiopathic testicular failure? (Results
NA, Miciek R, Krasnoff J, Elmi A, Bojesen A, Stockholm K, Juul S, of a double blind study). Hum
Knapp PE, Brooks B, Appleman E, Gravholt CH (2011) Socioeconomic Reprod 10:26002602
Aggarwal S, Bhasin G, Hede-Brierley trajectories affect mortality in Cunningham GR, Hirshkowitz M,
L, Bhatia A, Collins L, LeBrasseur N, Klinefelter syndrome. J Clin Korenman SG, Karacan I (1990)
Fiore LD, Bhasin S (2010) Adverse Endocrinol Metab 96:20982104 Testosterone replacement therapy
events associated with testosterone Brabrand S, Foss SD, Cvancarova M, and sleep-related erections in
administration. N Engl J Med Lehne G (2011) Androgen hypogonadal men. J Clin Endocrinol
363:109222 substitution with testosterone Metab 70:792797
Behre HM, Bohmeyer J, Nieschlag E undecanoate in survivors of bilateral Decker R, Pratsch C-J, Sippell WG
(1994) Prostate volume in testicular cancer requires (2002) Combined treatment with
testosterone-treated and untreated individually-adjusted injection testosterone (T) and ethinylestradiol
hypogonadal men in comparison intervals. BJU Int 107:10801087 (EE2) in constitutionally tall boys: is
to age-matched normal controls. Brown D, Butler CGE, Kelnar CJH, Wu treatment with T plus EE2 more
Clin Endocrinol 40:341349 FCW (1995) A double blind, effective in reducing final height in
Behre HM, Abshagen K, Oettel M, placebo controlled study of the tall boys than T alone? J Clin
Hbler D, Nieschlag E (1999a) effects of low dose testosterone Endocrinol Metab 87:16341639
Intramuscular injection of undecanoate on the growth of small de Lange WE, Snoep MC, Doorenbos H
testosterone undecanoate for the for age, prepubertal boys. Arch Dis (1979) The effect of short-term
treatment of male hypogonadism: Child 73:141145 testosterone treatment in boys with
phase I studies. Eur Bchter D, Behre HM, Kliesch S, delayed puberty. Acta Endocrinol
J Endocrinol 140:414419 Nieschlag E (1998) Pulsatile GnRH 91:177183
Behre HM, von Eckardstein S, Kliesch S, or human chorionic gonadotropin/ Depenbusch M, von Eckardstein S,
Nieschlag E (1999b) Long-term human menopausal gonadotropin Simoni M, Nieschlag E (2002)
substitution therapy of hypogonadal as effective treatment for men with Maintenance of spermatogenesis in
men with transscrotal testosterone hypogonadotropic hypogonadism: hypogonadotropic hypogonadal
over 710 years. Clin Endocrinol a review of 42 cases. Eur men with hCG alone. Eur
50:629635 J Endocrinol 149:298303 J Endocrinol 147:617624

305
Chapter 14: Clinical use of testosterone

de Ronde W (2009) Hyperandrogenism Ishii T, Sasaki G, Hasegawa T, Sato S, mesterolone in hypogonadal


after transfer of topical Matsuo N, Ogata T (2004) male patients. J Endocrinol Invest
testosterone gel: case report and Testosterone enanthate therapy is 3:305308
review of published and effective and independent of McNicholas T, Ong T (2006) Review of
unpublished studies. Hum Reprod SRD5A2 and AR gene Testim gel. Expert Opin
24:425428 polymorphisms in boys with Pharmacother 7:477484
de Waal WJ, Vreeburg JTM, Bekkering F, micropenis. J Urol 172:319324
Morales A, Johnston B, Heaton JPW,
de Jong FH, de Muinck Keizer- Jockenhvel F (1999) Influence of Lundie M (1997) Testosterone
Schrama SMPF, Drop SLS, Weber various modes of androgen supplementation for hypogonadal
RFA (1995) High dose testosterone substitution on serum lipids and impotence: assessment of
therapy for reduction of final height lipoproteins in hypogonadal men. biochemical measures and
in constitutionally tall boys: does it Metabolism 48:590596 therapeutic outcomes. J Urol
influence testicular function in Kamischke A, Nieschlag E (1999) 157:849854
adulthood? Clin Endocrinol Analysis of medical treatment
43:8795 Nieschlag E, Cppers HJ,
of male infertility. Hum Reprod Wiegelmann W, Wickings EJ (1976)
Drop SLS, de Waal WJ, de Muinck 14:123 Bioavailability and LH suppressing
Keizer-Schrama SMPF (1998) Sex
Khnert B, Byrne M, Simoni M, effect of different testosterone
steroid treatment of constitutionally
Kpcke W, Gerss J, Lemmnitz G, preparations in normal and
tall stature. Endocr Rev
Nieschlag E (2005) Testosterone hypogonadal men. Horm
19:540558
substitution with a new Res 7:148
Endres W, Shin YS, Rieth M, Block T, transdermal, hydroalcoholic gel Nieschlag E, Behre HM, Bouchard P,
Schmiedt E, Knorr D (1987) applied to scrotal or non-scrotal Corrales JJ, Jones TH, Stalla GK,
Priapism in Fabrys disease during skin: a multicentre trial. Eur Webb SM, Wu FC (2004)
testosterone treatment. Klin Wschr J Endocrinol 153:317326 Testosterone replacement therapy:
65:925
Lanfranco F, Kamischke A, Zitzmann current trends and future
Franchi F, Luisi M, Kicovic PM (1978) M, Nieschlag E (2004) Klinefelters directions. Hum Reprod Update
Long-term study of oral syndrome. Lancet 364:273283 10:409419
testosterone undecanoate in
hypogonadal males. Int J Androl Laughlin GA, Barrett-Connor E, Nieschlag E, Behre HM, Nieschlag S
1:270278 Bergstrom J (2008) Low serum (eds) (2010) Andrology. Male
testosterone and mortality in older Reproductive Health and
Gooren LJG (1994) A ten year safety men. J Clin Endocrinol Metab Dysfunction, 3rd edn. Springer-
study of the oral androgen 93:6875 Verlag, Berlin
testosterone undecanoate. J Androl
15:212215 Lee KK, Berman N, Alexander GM, Ohlsson C, Vandenput L (2009)
Hull L, Swerdloff RS, Wang C The role of estrogens for male
Haring R, Vlzke H, Steveling A, Krebs (2003) A simple self-report diary for bone health. Eur J Endocrinol
A, Felix SB, Schfl C, Drr M, assessing psychosexual function in 160:883889
Nauck M, Wallaschofski H (2010) hypogonadal men. J Androl
Low serum testosterone levels are Pedersen MF, Moller S, Krabbe S,
25:688698 Bennett P (1986) Fundamental
associated with increased risk of
mortality in a population-based Lemcke B, Zentgraf J, Behre HM, voice frequency measured by
cohort of men aged 2079. Kliesch S, Bramswig JH, Nieschlag E electroglottography during
Eur Heart J 31:14941501 (1996) Long-term effects on continuous speech. A new exact
testicular function of high-dose secondary sex characteristic in boys
Hendriks AE, Boellaard WP, van testosterone treatment for in puberty. Int J Ped Otohinol
Casteren NJ, Romijn JC, de Jong excessively tall stature. J Clin 11:2127
FH, Boot AM, Drop SL (2010) Endocrinol Metab 81:296301
Fatherhood in tall men treated with Pusch HH (1989) Oral treatment of
high-dose sex steroids during Luboshitzky R, Aviv A, Hefetz A, Herer P, oligozoospermia with testosterone-
adolescence. J Clin Endocrinol Shen-Orr Z, Lavie L, Lavie P (2002) undecanoate: results of a double-
Metab 95:52335240 Decreased pituitary-gonadal secretion blind-placebo-controlled trial.
in men with obstructive sleep apnea. Andrologia 21:7682
Ichioka K, Utsunomiya N, Kohei N,
J Clin Endocrinol Metab 87:33943398 Raynaud JP, Augs M, Liorzou L,
Ueda N, Inoue K, Terai A (2006)
Testosterone-induced priapism in Luisi M, Franchi F (1980) Double-blind Turlier V, Lauze C (2009)
Klinefelter syndrome. Urology group comparative study of Adhesiveness of a new testosterone-
67:622.e17622.e18 testosterone undecanoate and in adhesive matrix patch after

306
Chapter 14: Clinical use of testosterone

extreme conditions. Int J Pharmacol controlled study. Clin Endocrinol Wang C, Harnett M, Dobs AS,
375:2832 14:4961 Swerdloff RS (2010)
Rogol AD (2005) New facets of Snyder PJ (1999) Effect of testosterone Pharmacokinetics and safety of
androgen replacement therapy treatment on bone mineral density long-acting testosterone
during childhood and adolescence. in men over 65 years of age. J Clin undecanoate injections in
Expert Opin Pharmacother Endocrinol Metab 84:19661972 hypogonadal men: an 84-week
6:13191336 phase III clinical trial. J Androl
Snyder PJ, Lawrence DA (1980) 31:457465
Rolf C, von Eckardstein S, Koken U, Treatment of male hypogonadism
Nieschlag E (2002) Testosterone Warne DW, Decosterd G, Okada H,
with testosterone enanthate. J Clin
Yano Y, Koide N, Howles CM
substitution of hypogonadal men Endocrinol Metab 51:13351339
prevents the age-dependent (2009) A combined analysis of data
Sokol RZ, Palacios A, Campfield LA, to identify predictive factors for
increases in body mass index, body
Saul C, Swerdloff RS (1982) spermatogenesis in men with
fat and leptin seen in healthy ageing
Comparison of the kinetics of hypogonadotropic hypogonadism
men: results of a cross-sectional
injectable testosterone in eugonadal treated with recombinant human
study. Eur J Endocrinol 146:505511
and hypogonadal men. Fertil Steril follicle-stimulating hormones and
Rosen RC, Araujo AB, Connor MK, 37:425430 human chorionic gonadotropin.
Gerstenberger EP, Morgentaler A, Fertil Steril 92:594604
Seftel AD, Miner MM, Shabsigh R Vermeulen A, Verdonck G (1992)
(2011) The NERI Hypogonadism Representatives of a single point Whitworth JA, Scoggins BA, Andrews J,
Screener: psychometric validation plasma testosterone level for the Williamson PM, Brown MA
in male patients and controls. Clin long term hormonal milieu in men. (1992) Haemodynamic and
Endocrinol 74:248256 J Clin Endocrinol Metab 74:939942 metabolic effects of short term
von Eckardstein S, Nieschlag E (2002) administration of synthetic sex
Rosenfeld RG, Northcraft GB, steroids in humans. Clin Exp
Hintz RL (1982) A prospective, Treatment of male hypogonadism
with testosterone undecanoate Hypertens 14:905922
randomized study of testosterone
treatment of constitutional delay of injection at extended intervals of Wittert GA, Chapman IM, Haren MT,
growth and development in male 12 weeks: a phase II study. J Androl Mackintosh S, Coates P, Morley JE
adolescents. Pediatrics 69:681687 23:419425 (2003) Oral testosterone
Wang C, Alexander G, Berman N, supplementation increases muscle
Ruch W, Jenny P (1989) Priapism and decreases fat mass in healthy
following testosterone Salehian B, Davidson T, McDonald
V, Steiner B, Hull H, Callegari C, elderly males with low-normal
administration for delayed male gonadal status. J Gerontol A Biol Sci
puberty. Am J Med 86:256 Swerdloff R (1996) Testosterone
replacement therapy improves Med Sci 58:618625
Schulte-Beerbhl M, Nieschlag E (1980) mood in hypogonadal men a World Health Organization, Task
Comparison of testosterone, clinical research center study. J Clin Force on the Diagnosis and
dihydrotestosterone, luteinizing Endocrinol Metab 81: Treatment of Infertility (1989)
hormone, and follicle-stimulating 35783583 Mesterolone and idiopathic male
hormone in serum after injection of infertility: a double-blind study. Int
testosterone enanthate or testosterone Wang C, Cunningham G, Dobs A
J Androl 12:254264
cypionate. Fertil Steril 33:201203 (2004) Long-term testosterone gel
(AndroGel) treatment maintains World Health Organization, Nieschlag
Shabsigh R (1997) The effects of beneficial effects on sexual function E, Wang C, Handelsman DJ,
testosterone on the cavernous tissue and mood, lean and fat mass, and Swerdloff RS, Wu F, Einer-Jensen N,
and erectile function. World J Urol bone mineral density in Waites G (1992) Guidelines for the
15:2126 hypogonadal men. J Clin Endocrinol Use of Androgens. WHO, Geneva
Shores MM, Matsumoto AM, Sloan KL, Metab 89:20852098 Zelissen PMJ, Stricker BHC (1988)
Kivlajam DR (2006) Low serum Severe priapism as a complication
Wang C, Nieschlag E, Swerdloff R,
testosterone and mortality in of testosterone substitution therapy.
Behre HM, Hellstrom WJ, Gooren LJ,
male veterans. Arch Intern Med Am J Med 85:273
Kaufman JM, Legros JJ, Lunenfeld B,
166:16601665
Morales A, Morley JE, Schulman C, Zitzmann M, Nieschlag E (2001)
Skakkebaek NE, Bancroft J, Thompson IM, Weidner W, Wu FC Testosterone levels in healthy men
Davidson DW, Warner P (1981) (2008) Investigation, treatment and and their relation to behavioural
Androgen replacement with oral monitoring of late onset and physical characteristics: facts
testosterone undecanoate in hypogonadism in males. Eur and constructs. Eur J Endocrinol
hypogonadal men: a double blind J Endocrinol 159:507514 144:183197

307
Chapter 14: Clinical use of testosterone

Zitzmann M, Nieschlag E (2007) bone density in hypogonadal receptor gene: a longitudinal


Androgen receptor gene CAG men by quantitative phalangeal pharmacogenetic study. J Clin
repeat length and body mass index ultrasound. Bone 31: Endocrinol Metab 88:
modulate the safety of long-term 422429 20492054
intramuscular testosterone Zitzmann M, Depenbusch M, Gromoll J, Zitzmann M, Faber S, Nieschlag E
undecanoate therapy in Nieschlag E (2003) Prostate volume (2006) Association of specific
hypogonadal men. J Clin Endocrinol and growth in testosterone- symptoms and metabolic risks
Metab 92:38443853 substituted hypogonadal men are with serum testosterone in older
Zitzmann M, Rolf C, Brune M, Vieth V, dependent on the CAG repeat men. J Clin Endocrinol Metab
Nieschlag E (2002) Monitoring polymorphism of the androgen 91:43354343

308
Chapter
Testosterone preparations for clinical

15 use in males
Hermann M. Behre and Eberhard Nieschlag

15.1 Introduction 309 15.6.3 Testosterone cypionate and


15.2 Testosterone preparations for oral testosterone
administration 310 cyclohexanecarboxylate 318
15.2.1 Unmodified testosterone 310 15.6.4 Testosterone ester combinations 319
15.2.2 17a-Methyltestosterone 312 15.6.5 Testosterone buciclate 320
15.2.3 Fluoxymesterone 312 15.6.6 Testosterone undecanoate 321
15.2.4 Mesterolone 313 15.6.7 Testosterone decanoate 323
15.2.5 Testosterone undecanoate 313 15.6.8 Testosterone microcapsules 323
15.3 Testosterone preparations for sublingual or 15.7 Testosterone pellets for subdermal
buccal administration 314 administration 323
15.4 Testosterone preparations for nasal 15.8 Testosterone preparations for transdermal
administration 315 administration 324
15.5 Testosterone preparations for rectal 15.8.1 Testosterone patches 324
administration 315 15.8.2 Testosterone gels 325
15.6 Testosterone preparations for intramuscular 15.8.3 Testosterone topical
administration 315 solution 330
15.6.1 Testosterone propionate 316 15.9 Key messages 330
15.6.2 Testosterone enanthate 317 15.10 References 331

15.1 Introduction Today oral, buccal, injectable, implantable and trans-


Although testosterone has been in clinical use for almost dermal testosterone preparations are available for clin-
70 years, it has only slowly attracted interest from clinical ical use. There are only a few, especially few not-
researchers. This is partly due to the fact that hypogonadal industry-sponsored, clinical studies available comparing
men requiring testosterone treatment constitute only a the various preparations with the goal of identifying the
minority of all patients, and hypogonadism is not a life- optimal preparation for substitution purposes. While
threatening disease. Since development of new prepar- the older injectable testosterone preparations produce
ations is mainly a task of the pharmaceutical industry, and supraphysiological serum testosterone levels, newer
hypogonadal patients did not promise to contribute a preparations achieve levels closer to the physiological
substantial economic profit, development of testosterone range. We are only beginning to understand which
preparations was slow. Only recently has the question of serum levels are required to achieve the various bio-
testosterone treatment of senescent men (see Chapter 16) logical effects of testosterone and to avoid adverse side-
and, to a certain extent, also the search for a hormonal effects. In particular, very little is known about long-term
male contraceptive (see Chapter 22) spurred interest in effects of testosterone therapy inherent to different prep-
the pharmacology and application of testosterone. arations. Similarly, the role of the AR polymorphism in

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

309
Chapter 15: Testosterone preparations for clinical use

modifying testosterone action individually is becoming position 3, a hydroxy group in position 17 and a
understood only slowly, but may lead to a pharmacoge- double bond in position 4 (Fig. 15.1).
netic concept for the therapeutic application of testoster- Three approaches have been used to make testos-
one (Zitzmann 2009; see also Chapter 3). terone therapeutically effective: (1) various routes of
Under these circumstances it appears that the con- administration; (2) esterification in position 17; and
sensus reached by a Workshop Conference on Androgen (3) chemical modification of the molecule. In add-
Therapy organized jointly by the WHO, NIH and FDA ition, these approaches have been combined. Since it
in 1990 still provides the best therapeutic guidelines: The is of practical clinical relevance, the route of adminis-
consensus view was that the major goal of therapy is to tration is used here for categorizing the various con-
replace testosterone levels at as close to physiological ventional and new testosterone preparations for
concentrations as is possible (World Health Organiza- clinical use in males (overview in Table 15.1).
tion 1992). Until other evidence is provided, all testoster-
one preparations are best judged by this principle.
An important question is which androgen prepar- 15.2 Testosterone preparations
ation should be used for clinical purposes. Numerous for oral administration
androgenic steroids have been synthesized and used
clinically in the past. The synthetic androgens were 15.2.1 Unmodified testosterone
produced with the aim of selectively enhancing certain Unmodified testosterone as physiologically secreted by
aspects of testosterone activity, e.g. the anabolic effect the testes would appear to be the first choice when con-
on muscles or the hematopoietic effect. Some of these sidering substitution therapy. When ingested orally in its
molecules proved to have toxic side-effects, in particu- unmodified form, testosterone is absorbed well from the
lar upon long-term use (as required for substitution of gut but is effectively metabolized and inactivated in the
hypogonadism), or the desired efficacy and safety were liver before it reaches the target organs (first-pass effect).
inadequate in controlled clinical trials (as advocated by Only when a dose of 200 mg is ingested, which exceeds
evidence-based medicine) (see Chapter 25). In add- 30-fold the amount of testosterone produced daily by a
ition, some of these steroids cannot be converted to normal man, is the metabolizing capacity of the liver
5a-DHT or estrogen, as is testosterone, and therefore overcome. With such doses an increase in peripheral
cannot develop the full spectrum of activities of testos- testosterone blood levels becomes measurable and clinical
terone. The important biological significance of these effects can be observed (Johnsen et al. 1974; Nieschlag
conversions is described in Chapter 2. For these et al. 1975). The testosterone-metabolizing capacity of the
reasons, synthetic preparations have almost disap- liver, however, is age- and sex-dependent. An oral dose of
peared from the market, and testosterone as produced 60 mg unmodified testosterone does not affect peripheral
naturally is the prevailing androgen used in clinical testosterone levels in normal adult men, but produces a
medicine. In its various preparations testosterone has significant rise in prepubertal boys and women (Nieschlag
been available for over seven decades and, as one of the et al. 1977). This demonstrates that testosterone induces
oldest drugs in clinical use, has demonstrated its high liver enzymes responsible for its own metabolism (John-
safety. However, new insights into the molecular sen et al. 1976). When the liver is severely damaged its
mechanisms of androgen action may lead to the devel- metabolizing capacity decreases. Thus, in patients with
opment of steroids suited for specific purposes (see liver cirrhosis a dose of 60 mg testosterone (ineffective
Chapter 21). Whether such steroids may become in normal men) produces high serum levels (Nieschlag
useful and safe for clinical use remains to be seen. et al. 1977).
As all other androgens, testosterone derives from In hypogonadal men with normal liver function,
the basic structure of androstane. This molecule con- 400600 mg testosterone must be administered daily
sists of three cyclohexane and one cyclopentane ring if the patient is to be substituted by oral testosterone
(perhydrocyclopentanephenanthrene ring) and a (Johnsen et al. 1974) a dose exceeding the testoster-
methyl group each in position 10 and 13. Androstane one production of a normal man almost 100-fold.
itself is biologically inactive and gains activity through Aside from being uneconomical, the possibility of
oxygroups in position 3 and 17. Testosterone, the adverse effects of such huge testosterone doses cannot
quantitatively most important androgen synthesized be excluded, especially when given over long periods
in the organism, is characterized by an oxy group in of time as required for substitution therapy.

310
Chapter 15: Testosterone preparations for clinical use

OH OCOCH2CH3

Testosterone Testosterone
O O propionate

OH OCO(CH2)5CH3
CH3

17-Methyltestosterone Testosterone
O O enanthate

OH OCO(CH2)2

HO CH3

Fluoxymesterone Testosterone
O O cypionate

OH OCO (CH2)3CH3

CH3

Mesterolone Testosterone butyl


O O cyclohexylcarboxylate

OCO(CH2)2 O(CH2)5CH3 OCO(CH2)9CH3


CH3

19Nortestosterone Testosterone
O hexoxyphenylpropionate O undecanoate
Fig. 15.1 Molecular structure of testosterone and clinically used testosterone esters and derivatives.

Recently, administration of high doses of testos- to the normal or supraphysiological range. In addition,
terone has been re-examined with and without con- three-times 300 mg of oral testosterone per day was
comitant inhibition of 5a-reductase by dutasteride or tested in normal men with suppressed endogenous
finasteride (Snyder et al. 2010; Amory et al. 2011). testosterone by GnRH antagonist administration (Lee
Administration of 150400 mg oral testosterone in et al. 2011). Long-term studies with data on potential
various formulations twice daily or 300600 mg daily side-effects of these high-dose testosterone prepar-
with and without concomitant 5a-reductase inhibi- ations and the clinical relevance of significantly
tors to hypogonadal men or men with experimental decreased DHT levels are awaited to estimate the clin-
hypogonadism increased serum levels of testosterone ical perspective of this therapeutic approach.

311
Chapter 15: Testosterone preparations for clinical use

Table 15.1 Mode of application and dosage of various testosterone preparations

Preparation Route of application Full substitution dose


In clinical use
Testosterone undecanoate Oral 24 capsules at 40 mg/day
Testosterone tablets Buccal 30 mg twice daily
Testosterone enanthate Intramuscular injection 200250 mg every 23 weeks
Testosterone cypionate Intramuscular injection 200 mg every 2 weeks
Testosterone undecanoate Intramuscular injection 1000 mg every 1014 weeks
Testosterone implants Implantation under the 4 implants at 200 mg every 56 months
abdominal skin
Transdermal testosterone patch Non-scrotal skin 2 systems per day or every second day
depending on preparation
Transdermal testosterone gel (12.5%) Non-scrotal skin Starting dose 4062.5 mg/day depending
on preparation
Testosterone solution (2%) Axillary Starting dose 60 mg/day
In advanced clinical development
Testosterone undecanoate (self-emulsifying Oral Not yet determined
drug delivery system)
Obsolete
17a-Methyltestosterone Oral
Fluoxymesterone Sublingual/oral

15.2.2 17a-Methyltestosterone treatment and liver tumors was found (e.g. Farrell
et al. 1975; Boyd and Mark 1977; Goodman and
Several attempts have been made to modify the testos-
Laden 1977; Paradinas et al. 1977; Falk et al. 1979).
terone molecule by chemical means in order to render
The hepatotoxic side-effects are due to the alkyl
it orally effective; i.e. to delay metabolism in the liver.
group in the 17a position, and have also been reported
In this regard, the longest-known testosterone deriva-
for other steroids with this configuration (Krskemper
tive is 17a-methyltestosterone (Ruzicka et al. 1935),
and Noell 1967). Because of the side-effects methyltes-
which is a fully effective oral androgen preparation.
tosterone should no longer be used therapeutically for
17a-Methyltestosterone is quickly absorbed, and max-
hypogonadism, in particular since effective alterna-
imal blood levels are observed 90 to 120 minutes after
tives are available (Nieschlag 1981). The German
ingestion. The half-life in blood amounts to approxi-
Endocrine Society declared methyltestosterone obso-
mately 150 minutes (Alkalay et al. 1973).
lete in 1981, and the German Federal Health Authority
Ever since this steroid was introduced for clinical
ruled that methyltestosterone should be withdrawn
use, hepatotoxic side-effects such as an increase in
from the market (Anonymous 1988). In other coun-
serum liver enzymes (Carbone et al. 1959), cholestasis
tries, however, methyltestosterone is still in use, a
of the liver (Werner et al. 1950; de Lorimer et al.
practice which should be terminated.
1965) and peliosis of the liver (Westaby et al. 1977)
have been reported repeatedly. It is of interest that
humans are more susceptible to the hepatotoxic 15.2.3 Fluoxymesterone
effects of methyltestosterone than rats (Heywood The androgenic activity of fluoxymesterone was
et al. 1977a) or dogs (Heywood et al. 1977b). Later, enhanced over that of testosterone by the introduc-
an association between long-term methyltestosterone tion of fluorine and the addition of a hydroxy group

312
Chapter 15: Testosterone preparations for clinical use

into the steroid skeleton of testosterone. This sub- 60


stance also contains a 17a-methyl group, and accord-
ingly there is a risk of hepatotoxicity with long-term 50

Testosterone (nmol / l)
use. Therefore, this androgen has disappeared from 40
the market.
30
15.2.4 Mesterolone 20
Mesterolone can be considered a derivative of the 5a-
reduced testosterone metabolite 5a-dihydrotestoster- 10
one (DHT), which is protected from rapid metabol-
ism in the liver by a methyl group in position 1 0
Basal 4 3 2 1 0 1 2 3 4 5
(Gerhards et al. 1966) and thus becomes orally active. Time (h)
It is free of liver toxicity. Unlike testosterone, mester-
Fig. 15.2 Single-dose pharmacokinetics of testosterone
olone cannot be metabolized to estrogens (Breuer and undecanoate dissolved in oleic acid after oral administration of
Gtgemann 1966), and at a molecular level acts like 120 mg of the ester to eight hypogonadal patients. Because of high
DHT. Because of its limited effectiveness in suppress- interindividual variability of testosterone serum concentrations after
administration of testosterone undecanoate, individual curves were
ing pituitary gonadotropin secretion (Aakvaag and all centralized about the time of maximal serum concentrations
Stromme 1974; Gordon et al. 1975), it can only be (time 0). Asterisks indicate significant higher testosterone serum
considered an incomplete androgen. Altogether, mes- concentrations compared to pretreatment values (basal) (mean 
standard error of mean (SEM)).
terolone is not suited for the substitution of
hypogonadism.
versus time curves were centralized about the time
15.2.5 Testosterone undecanoate of maximal serum concentrations, serum concentra-
When testosterone is esterified in the 17b-position tions significantly different from basal values were
with a long fatty acid side chain such as undecanoic seen only two hours before and one hour after the
acid, and given orally, its route of absorption from the time of maximal serum concentrations in hypogona-
gastrointestinal tract is slightly shifted from the vena dal patients (Fig. 15.2; Schrmeyer et al. 1983). Based
portae to the lymph and reaches the circulation via on this observation it can be deduced that, even with
the thoracic duct (Coert et al. 1975; Horst et al. 1976; administration of testosterone undecanoate in oleic
Shackleford et al. 2003). Absorption is improved if the acid three times daily, only short-lived testosterone
ester is taken in arachis oil (Nieschlag et al. 1975) and peaks resulting in high fluctuations can be obtained.
with a meal (Frey et al. 1979; Bagchus et al. 2003). The initial commercial preparation of oral testos-
After oral ingestion of a 40 mg capsule, of which 63%, terone undecanoate in oleic acid had to be refriger-
i.e. 25 mg, is testosterone, maximum serum levels are ated (28  C) in the pharmacy for reasons of stability;
reached two to six hours later (Nieschlag et al. 1975). whereas patients had to store it at room temperature
Thus, with two to four capsules (80 to 160 mg) per to ensure optimal absorption. The shelf-life at room
day, testosterone substitution of hypogonadism can temperature was only three months. Therefore, a new,
be achieved. more stable pharmaceutical formulation of testoster-
Pharmacokinetics of testosterone undecanoate one undecanoate was developed in which the oleic
after single-dose administration in oleic acid was acid was replaced by castor oil. This new formulation
tested in 8 hypogonadal patients and 12 normal men can be stored at room temperature for three years.
(Schrmeyer et al. 1983). On average, maximum The lipid content in food influences absorption of this
levels of serum testosterone could be observed five oral testosterone undecanoate preparation and could
hours after testosterone undecanoate administration. explain some of the intraindividual variability of tes-
However, the serum testosterone profile showed high tosterone levels. A low liquid meal with, at most, 5 g
interindividual variability of the time when maximum of lipid is not sufficient for adequate absorption of
concentrations were reached, as well as of the max- testosterone undecanoate. However, a normal meal
imum levels themselves, which ranged from 17 to 96 containing approximately 19 g of lipid efficiently
nmol/l. When the individual serum concentration increases serum testosterone levels after oral

313
Chapter 15: Testosterone preparations for clinical use

administration of testosterone undecanoate capsules (A)


(Schnabel et al. 2007).
Recently, testosterone undecanoate was formu-
lated into a new proprietary self-emulsifying drug
delivery system (SEDDS) preparation for oral applica-
tion. This SEDDS is intended to promote solubiliza-
tion and the intestinal lymphatic absorption of
lipophilic testosterone esters and thereby reduce first-
pass hepatic metabolism. Pharmacokinetic studies in a.m. dose
hypogonadal men up to 28 days showed that 200 mg
testosterone twice daily given as testosterone unde-
canoate SEDDS (100 mg testosterone = 158.3 mg tes-
tosterone undecanoate) increased mean serum levels
of testosterone to the normal range in most subjects,

Testosterone (nmol/I)
but not when taken in the fasting state (Yin et al. 2012;
Fig. 15.3). Currently, this testosterone preparation is
being tested in a large international phase III study.
(B)
15.3 Testosterone preparations for
sublingual or buccal administration
17a-Methyltestosterone was found to be more effect-
ive when applied sublingually than when ingested
orally (Escamilla 1949). This type of substitution
should, however, not be practiced, because of the liver
toxicity of methyltestosterone summarized above.
The solubility of the hydrophobic testosterone
molecule can be enhanced by incorporation into
hydroxypropyl-b-cyclodextrins (Pitha et al. 1986),
which are macro-ring structures consisting of cyclic
oligosaccharides. When testosterone incorporated
into such cyclodextrins is administered sublingually, a.m. dose p.m. dose
steep increases in serum testosterone occur, lasting
for one or two hours (Stuenkel et al. 1991). Hypogo-
nadal men treated with three-times-daily doses for 60
Time (h)
days showed improvement of their condition (Sale-
hian et al. 1995; Wang et al. 1996). This is an interest- Fig. 15.3 Pharmacokinetics of testosterone undecanoate
formulated into a proprietary self-emulsifying drug delivery system
ing approach to testosterone substitution, but unless (SEDDS) (mean  SEM) (adapted with permission from Yin et al.
more constant serum levels can be achieved this ther- 2012). (A) Single dose pharmacokinetics after oral dosing of 200 mg
apy would require repeated daily applications and testosterone in 12 hypogonadal men (100 mg testosterone =
158.3 mg testosterone undecanoate). (B) Multiple dose
would have the same disadvantages as conventional pharmacokinetics on day seven after 300 mg testosterone
oral testosterone undecanoate therapy. administered twice daily for seven days.
Administration of testosterone via the buccal
mucosa bypasses the liver and avoids first-pass clear- testosterone bioadhesive buccal system was designed
ance by delivering the drug directly into systemic to adhere rapidly to the buccal mucosa and gellify, for
circulation. Compared to sublingual administration, delivering testosterone steadily into the circulation.
buccal mucosa is less permeable and potentially better The pharmacokinetics were evaluated in 82 hypogo-
suited for sustained delivery systems. nadal men (Wang et al. 2004). The tablet (30 mg
Whereas earlier buccal testosterone preparations testosterone) was applied twice daily to the upper
were not very acceptable to the patients, a new gums for three months; 86.6% of the patients reached

314
Chapter 15: Testosterone preparations for clinical use

an average testosterone concentration over 24 hours would have to be injected very frequently. Esterifica-
within the physiological range. Local problems asso- tion of the testosterone molecule at position 17, for
ciated with tablet use were transient in most patients. example, with propionic or enanthic acid, prolongs
Long-term acceptability and compliance of this mode the activity of testosterone in proportion to the length
of testosterone delivery to hypogonadal patients has of the side chain when administered intramuscularly
still to be awaited. For example, in Germany this (Junkmann 1952; 1957). The deep im injection of
preparation was launched some years ago for treat- testosterone esters in oily vehicle is generally safe
ment of hypogonadism but has disappeared from the and well tolerated, but can cause minor side-effects
market in the meantime. such as local pain (Mackey et al. 1995).
Studies applying GC-MS that allow discrimination
15.4 Testosterone preparations for between endogenous testosterone and exogenously
administered deuterium-labeled testosterone
nasal administration propionate-19,19,19-d3 and its metabolite testoster-
The first-pass effect of the liver can also be avoided by one-19,19,19-d3 were able to show that after im
applying testosterone to the nasal mucosa (Danner administration, the testosterone ester is slowly absorbed
and Frick 1980). However, unreliable absorption into the general circulation and then rapidly converted
patterns and short-lived serum peaks prevented this to the active unesterified metabolite (Fujioka et al.
form of application from further development to 1986). The observation that the duration at the injection
become a desirable option for long-term substitution site is the major factor determining the residence time
therapy. Recently, this form of application has been of the drug in the body agrees with pharmacokinetic
re-examined: 7.6 mg of nasal testosterone was given studies in rats showing that the androgen ester
either twice or three times a day in 21 severely hypo- 19-nortestosterone decanoate, when injected into the
gonadal men for 14 days (Mattern et al. 2008). In musculus gastrocnemius of the rat in vivo, is absorbed
the group treated three times a day, serum levels of unchanged from the injection depot in the muscle into
testosterone were maintained in most patients within the general circulation according to first-order kinetics,
the normal range. Further long-term studies will be with a long half-life of 130 h (van der Vies 1965).
necessary to fully evaluate the potential of this mode Comparisons of the absorption kinetics of different
of testosterone application. testosterone esters clearly show that the half-lives of
the absorption of the esters increase when the esterified
15.5 Testosterone preparations for fatty acids have a longer chain (van der Vies 1985).
In addition, pharmacokinetics are influenced by the oily
rectal administration vehicle, the injection site and the injection volume
In order to avoid the first-pass effect of the liver, (Minto et al. 1997).
testosterone can be applied rectally in suppositories After absorption from the intramuscular depot,
(Hamburger 1958). Administration of a suppository the testosterone ester is rapidly hydrolyzed in plasma,
containing 40 mg testosterone results in an immedi- as was shown by in-vitro rat studies (van der Vies
ate and steep rise of serum testosterone lasting for 1970) and in-vivo human studies (Fujioka et al. 1986).
about four hours. Effective serum levels can be The rate of hydrolysis again depends on the structure
achieved by repeated applications (Nieschlag et al. of the acid chain, but this process is much faster than
1976). This therapy, however, never gained much release from the injection depot (van der Vies 1985).
popularity probably because the patients find it The metabolism of the testosterone ester to the unes-
unacceptable to use suppositories three times daily terified testosterone occurs rapidly, so that testoster-
on a long-term routine basis. one enanthate or testosterone have nearly identical
intravenous pharmacokinetics (Sokol and Swerdloff
15.6 Testosterone preparations for 1986). Similarly, the duration of action of the orally
effective ester testosterone undecanoate seems to be
intramuscular administration dependent on the duration of absorption of the
The most widely used testosterone substitution ther- uncleaved lipophilic testosterone undecanoate via
apy is the im injection of testosterone esters. Unmodi- the ductus thoracicus from the gut (Maisey et al.
fied testosterone has a half-life of only 10 minutes and 1981; Schrmeyer et al. 1983).

315
Chapter 15: Testosterone preparations for clinical use

In men treated with testosterone, the testosterone Table 15.2 Comparative pharmacokinetics of different
testosterone esters after intramuscular injection to hypogonadal
concentration measurable in the serum is the sum of patients
endogenous testosterone and exogenous testosterone
hydrolyzed from the injected ester. Hypogonadal Testosterone ester Terminal elimination
patients are characterized by impaired or absent half-life (d)
endogenous testosterone secretion; exogenous testos- Testosterone 0.8
terone administration can further suppress endogen- propionate
ous testosterone secretion only to a limited degree, if
Testosterone 4.5
at all. Accordingly, in hypogonadal patients the serum enanthate
concentration versus time profile is mainly a reflec-
tion of the pharmacokinetics of exogenously adminis- Testosterone buciclate 29.5
tered testosterone ester alone. In this chapter the Testosterone 33.9
evaluation of pharmacokinetic parameters for differ- undecanoate
ent testosterone esters is based on the increases of
testosterone serum concentrations over basal levels
in hypogonadal patients. 50

40

Testosterone (nmol / l)
15.6.1 Testosterone propionate
Single-dose pharmacokinetics of 50 mg testosterone 30
propionate after im injection to seven hypogonadal
patients, and the best-fit pharmacokinetic profile, are 20
shown in Fig. 15.4 (Nieschlag et al. 1976). Maximal
testosterone levels in the supraphysiological range 10
were seen shortly after injection (40.2 nmol/l, tmax =
14 h). Testosterone levels below the normal range were 0
observed following day two (57 h) after injection. The 0 1 2 3 4 5 6
calculated values were 1843 nmolh/l for area under the Time (d)
curve (AUC); 1.5 d for mean residence time (MRT); Fig. 15.4 Single-dose pharmacokinetics of testosterone
and 0.8 d for terminal half-life (Table 15.2). propionate in seven hypogonadal patients. Closed circles, mean 
Based on single-dose pharmacokinetic param- SEM of testosterone serum concentrations actually measured; curve,
best-fit pharmacokinetic profile.
eters, a multiple-dose pharmacokinetic simulation
was performed. Expected testosterone serum concen-
trations after multiple dosing of 50 mg testosterone
propionate, twice per week (e.g. injections Mondays 50
and Thursdays, 8 a.m.), are shown in Fig. 15.5.
Shortly after injection, high supraphysiological testos- 40
Testosterone (nmol/I)

terone serum concentrations of up to 45 nmol/l are


observed. At the end of the injection interval (three 30
and four days, respectively), testosterone serum con-
centrations below the lower range of normal testoster- 20
one values are projected (7 nmol/l and 3 nmol/l,
respectively). 10
Judged by the data from pharmacokinetic analysis
and simulation, administration of testosterone propi- 0
0 2 4 6 8 10 12 14 16 18 20
onate is not suitable for substitution therapy of male
Time (d)
hypogonadism because of its short-term kinetics
resulting in wide fluctuations of testosterone serum Fig. 15.5 Multiple-dose pharmacokinetics of testosterone
propionate after injection of 50 mg testosterone propionate twice
concentrations and maximal injection intervals of per week (e.g. Mondays and Thursdays). Solid curve, pharmacokinetic
three days for the 50 mg dose. simulation; broken lines, range of normal testosterone values.

316
Chapter 15: Testosterone preparations for clinical use

15.6.2 Testosterone enanthate minimal testosterone serum concentrations up to


40 nmol/l just before the next injection (Fig. 15.7).
Single-dose pharmacokinetics of testosterone
Injecting 250 mg of testosterone enanthate every two
enanthate after im administration of 250 mg testoster-
weeks results in maximal supraphysiological
one enanthate to seven hypogonadal patients and the
best-fit pharmacokinetic profile are shown in Fig. 15.6 50
(Nieschlag et al. 1976). Maximal testosterone levels in

Testosterone (nmol/l)
the supraphysiological range were seen shortly after 40
injection (39.4 nmol/l, tmax = 10 h). Testosterone levels
below the normal range were observed following day 30
12 after injection. The calculated values were 9911
nmolh/l for AUC, 8.5 d for MRT and 4.5 d for ter- 20
minal half-life (Table 15.2).
Based on the pharmacokinetic parameters of 10
single-dose pharmacokinetics, multiple-dose pharma-
cokinetic simulations for equal doses of 250 mg testos- 0
0 5 10 15 20
terone enanthate and injection intervals of one to four
weeks were performed. With weekly injection inter- Time (d)
vals, supraphysiological maximal testosterone serum Fig. 15.6 Single-dose pharmacokinetics of testosterone enanthate
concentrations up to 78 nmol/l are observed at steady in seven hypogonadal patients. Closed circles, mean  SEM of
testosterone serum concentrations actually measured; curve, best-fit
state shortly after injection, and supraphysiological pharmacokinetic profile.

(A) Injection interval Fig. 15.7 Multiple-dose


1 week pharmacokinetics of testosterone
80 enanthate after injection of 250 mg
testosterone enanthate every week (A),
60 every second week (B), every three weeks
40 (C) and every four weeks (D). Solid curves,
pharmacokinetic simulations; broken lines,
20 range of normal testosterone values.
0

(B) 80
2 weeks
60
40
Testosterone (nmoI/I)

20
0

(C) 80
3 weeks
60
40
20
0

(D) 80
4 weeks
60
40
20
0
0 2 4 6 8 10 12 14 16
Time (weeks)

317
Chapter 15: Testosterone preparations for clinical use

testosterone serum concentrations of up to 51 nmol/l


shortly after injection and testosterone serum levels at 50

Testosterone (nmol/l)
the lower range for normal testosterone serum concen-
40
tration shortly before the next injection. If the injection
interval is extended to three weeks, testosterone serum 30
concentrations below the normal range are observed 14
days after injection. With injection intervals of four 20
weeks, testosterone serum concentrations are in the
subnormal range at week three and four, and effective 10
testosterone substitution is not guaranteed (Fig. 15.7).
The calculated testosterone serum concentrations 0
0 2 4 6 8 10
at steady state obtained by computer simulation Time (d)
correspond well to the results of published studies
Fig. 15.8 Comparative pharmacokinetics of 194 mg of
describing multiple-dose testosterone enanthate testosterone enanthate and 200 mg of testosterone cypionate after
pharmacokinetics. In a clinical trial for male contra- im injection to six normal volunteers. , mean  SEM of testosterone
ception, 20 healthy men were injected with 200 mg/wk levels after testosterone enanthate injections; , mean  SEM of
testosterone levels after testosterone cypionate injections.
of testosterone enanthate for 12 weeks (Cunningham
et al. 1978). Minimal serum concentrations of testos-
terone at steady state, i.e. the testosterone serum con- eventually reaching the lower normal limit (Snyder
centration just before the next injection, were and Lawrence 1980). Similar results were described
measured at 31.2 to 39.5 nmol/l after weekly injection after injection of 300 mg/3 wks or 400 mg/4 wks tes-
of 200 mg testosterone enanthate. Very similar data tosterone enanthate. The authors conclude that the
were obtained in further contraceptive studies when testosterone enanthate doses of 200 mg have to be
normal men received 200 mg/wk testosterone injected every two weeks or doses of 300 mg every
enanthate injections for 18 months (Anderson and three weeks to guarantee effective substitution therapy.
Wu 1996; Wu et al. 1996). The data from these studies
fit well with the computer-calculated minimal testos- 15.6.3 Testosterone cypionate and
terone serum concentrations of 40 nmol/l and max-
imal testosterone levels of 78 nmol/l after multiple testosterone cyclohexanecarboxylate
injections of testosterone enanthate at a dosage of Testosterone cypionate (cyclopentylpropionate) phar-
250 mg/wk. macokinetics were compared with those of testosterone
Snyder and Lawrence (1980) administered 100 mg/wk enanthate in a crossover study involving six healthy
(n = 12), 200 mg/2 wks (n = 10), 300 mg/3 wks (n = 9) men aged 2029 years. Three subjects received 194 mg
and 400 mg/4 wks (n = 6) testosterone enanthate to of testosterone enanthate, followed seven weeks later by
hypogonadal patients during a study period of three 200 mg of testosterone cypionate and vice versa
months. Blood was drawn during the last injection (amount of unesterified testosterone 140 mg in both
period, when steady state had been reached, every preparations). The serum testosterone profiles were
day (100 mg/wk) up to every fourth day (400 mg/4 identical after injection of both preparations in equiva-
wks). Similar to the computer simulation described lent doses, both in terms of maximal concentrations
above for 250 mg testosterone enanthate and injec- and in terms of duration of elevation above basal levels
tion intervals of one to four weeks, initial supraphy- (Fig. 15.8; Schulte-Beerbhl and Nieschlag 1980).
siological testosterone serum levels were seen shortly In a subsequent clinical study, the pharmacoki-
after injection. In the 100 mg/wk treatment group, netics of testosterone cyclohexanecarboxylate were
where daily blood sampling was performed, mean compared to the pharmacokinetics of testosterone
peak serum concentrations were seen 24 h after enanthate in a single-blind, crossover study in seven
injection. Comparable to the results of the computer healthy young men (Schrmeyer and Nieschlag
simulation, after injection of 200 mg/2 wks testos- 1984). After injection of either testosterone enanthate
terone enanthate, following initial supraphysio- or testosterone cyclohexanecarboxylate, testosterone
logical testosterone serum levels, values fell to concentrations in serum increased sharply and
progressively lower values before the next injection, reached maximum levels, four to five times above

318
Chapter 15: Testosterone preparations for clinical use

basal, 824 h after injection. During the following propionate and 55 mg testosterone enanthate; Tes-
days a parallel decay of testosterone levels occurred toviron Depot 100: 25 mg testosterone propionate
after injection of either ester preparation, with testos- and 110 mg testosterone enanthate; Sustanon 250:
terone serum concentrations slightly, but significantly 30 mg testosterone propionate, 60 mg testosterone
lower after testosterone cyclohexanecarboxylate injec- phenylpropionate, 60 mg testosterone isocaproate
tion compared to testosterone enanthate injection and 100 mg testosterone decanoate). These com-
two, three and seven days after administration. Basal binations are used following the postulate that the
serum levels were reached seven days after testoster- so-called short-acting testosterone ester (e.g. testos-
one cyclohexanecarboxylate administration and nine terone propionate) is the effective testosterone for
days after injection of testosterone enanthate. substitution during the first days of treatment, and
Because testosterone cypionate, testosterone the so-called long-acting testosterone (e.g. testos-
cyclohexanecarboxylate and testosterone enanthate terone enanthate) warrants effective substitution
had comparable suppressing effects on LH and con- for the end of the injection interval. However, this
sequently on endogenous testosterone secretion, it assumption is not supported by the pharmacoki-
can be concluded from these studies in normal vol- netic parameters of the individual testosterone
unteers that all three esters with similar molecular esters. Both testosterone propionate and testoster-
structure possess comparable pharmacokinetics of one enanthate cause highest testosterone serum
exogenous testosterone serum concentrations. Testos- concentrations shortly after injection (Fig. 15.4
terone cypionate or testosterone cyclohexanecarbox- and Fig. 15.6). Accordingly, addition of testoster-
ylate do not provide a more advantageous one propionate to testosterone enanthate
pharmacokinetic profile than testosterone enanthate. only increases the initial undesired testosterone
This observation is in agreement with a clinical study peak and worsens the pharmacokinetic profile that
of replacement therapy with single-dose administra- ideally should follow zero-order kinetics (Fig. 15.9).
tion of 200 mg of testosterone cypionate in 11 hypo- The computer simulation agrees well with the
gonadal patients (Nankin 1987). limited published single-dose testosterone values
that have been measured in hypogonadal patients
treated with the combination of testosterone propi-
15.6.4 Testosterone ester combinations onate and testosterone enanthate. Maximal
Testosterone ester mixtures have been widely used increases of approximately 40 nmol/l testosterone
for substitution therapy of male hypogonadism over basal values are described one day after im
(e.g. Testoviron Depot 50: 20 mg testosterone administration of a testosterone ester combination

40 Fig. 15.9 Pharmacokinetic profile of


Testoviron Depot 100 (110 mg
testosterone enanthate (TE) and 25 mg
Testoviron Depot 100 testosterone propionate (TP)) in
comparison to the pharmacokinetics of
30 the individual testosterone esters of the
mixture. Curves, pharmacokinetic
Testosterone (nmol/l)

simulations.

25 mg TP
20

110 mg TE
10

0
0 1 2 3 4 5 6 7 8
Time (d)

319
Chapter 15: Testosterone preparations for clinical use

(A) Fig. 15.10 Multiple-dose


50 pharmacokinetics of (A) the testosterone
ester mixture Testoviron Depot 100 (110
mg testosterone enanthate and 25 mg
40 testosterone propionate = 100 mg
unesterified testosterone) every 10 d, in
comparison with (B) 139 mg testosterone
30 enanthate (= 100 mg unesterified
testosterone) every 10 d. Solid curves,
20 pharmacokinetic simulations; broken lines,
range of normal testosterone values.

10
Testosterone (nmol/l)

(B)
50

40

30

20

10

0
0 10 20 30 40 50 60 70 80
Time (d)

of 115.7 mg testosterone enanthate and 20 mg


testosterone propionate to three hypogonadal 15.6.5 Testosterone buciclate
patients (Fukutani et al. 1974). The disadvantage of all esters described so far is that
A comparison of computer-simulated testosterone they produce initially supraphysiological testosterone
serum concentrations after multiple-dose injections of levels, which may exceed normal levels several-fold,
Testoviron Depot 100 (110 mg testosterone enanthate and then slowly decline, so that before the next
and 25 mg testosterone propionate = 100 mg unester- injection pathologically low levels may be reached.
ified testosterone) every 10 d, and 139 mg testosterone Some patients recognize these ups and downs of
enanthate (= 100 mg unesterified testosterone) every testosterone levels in parallel variations of general
10 d is shown in Fig. 15.10. As can be expected from the well-being, sexual activity and emotional stability.
single-dose kinetics of the individual esters, injection Despite these disadvantages, testosterone enanthate
of the testosterone ester mixture (Fig. 15.10A) pro- and cypionate are still the standard intramuscular
duces a much wider fluctuation of testosterone serum therapy for male hypogonadism in various countries.
concentrations relative to injection of testosterone Because of these shortcomings of the available
enanthate alone (Fig. 15.10B). This simulation shows esters, the WHO initiated a steroid synthesis program
that injections of testosterone enanthate alone produce (Crabb et al. 1980), out of which a series of new
a more favorable pharmacokinetic profile in compari- testosterone esters was developed. When tested in
son to injections of testosterone propionate and testos- laboratory rodents, a specific ester was identified that
terone enanthate ester mixtures in comparable doses. showed greatly prolonged activity, namely testoster-
For treatment of male hypogonadism there is no one-trans-4-n-butylcyclohexyl-carboxylate, generic
advantage in combining the available short- and name testosterone buciclate. This preparation is
long-acting testosterone esters. injected intramuscularly in an aqueous solution, in

320
Chapter 15: Testosterone preparations for clinical use

7 injected dose of testosterone buciclate from 600 to


1000 mg prolongs the duration of action significantly,
6
but does not lead to significantly higher maximal
Testosterone (nmol/l)

5 serum levels of testosterone.


The long duration of action of testosterone buci-
4
clate was also demonstrated in a contraceptive study
3 with this new testosterone ester. After a single im
injection of 1200 mg testosterone buciclate at a con-
2
centration of 400 mg/ml to eight normal men, serum
1 levels of testosterone remained within the normal
range, whereas gonadotropins and spermatogenesis
0
0 7 14 21 28 35 42 49 56 63 70 77 84 were significantly suppressed for at least 18 weeks
Time (d) (Behre et al. 1995). These studies demonstrate that
Fig. 15.11 Single-dose pharmacokinetics of testosterone buciclate
the long-acting testosterone buciclate is well suited for
after im injection of 600 mg of the ester to four hypogonadal substitution therapy of male hypogonadism as well as
patients. Closed circles, mean  SEM of testosterone serum for male contraception. However, this compound has
concentrations actually measured (increase over baseline); curve,
best-fit pharmacokinetic profile.
not been developed into a marketable product and is
currently not available.

contrast to the other testosterone esters which are


dissolved in oily solution. 15.6.6 Testosterone undecanoate
To assess the pharmacokinetics of testosterone While testosterone undecanoate has been available
buciclate in men, the first clinical study was per- for oral substitution for more than three decades, it
formed in eight men with primary hypogonadism was first demonstrated in China that im administra-
(Behre and Nieschlag 1992). The men were randomly tion of testosterone undecanoate in tea seed oil
assigned to two study groups and were given either (125 mg/ml) has a prolonged duration of action
200 mg (group I) or 600 mg (group II) testosterone (Wang et al. 1991). In a clinical study in Asian hypo-
buciclate, intramuscularly. Whereas in group I serum gonadal men, eight patients received one im injection
androgen levels did not rise to normal values, in of 500 mg, and seven of the initial eight hypogonadal
group II androgens increased significantly and were patients one injection of 1000 mg testosterone unde-
maintained in the normal range up to 12 weeks, with canoate (in 8 ml tea seed oil) in a crossover design
maximal serum levels (cmax) of 13.1  0.9 nmol/l (Zhang et al. 1998). Follow-up blood samples were
(mean  SEM) in study week six (tmax). No initial obtained weekly up to week nine after injection. In
burst release of testosterone was observed in either both study groups, mean serum levels of testosterone
study group. Pharmacokinetic analysis revealed a were above the upper limit of normal during the first
terminal elimination half-life of 29.5  3.9 days two weeks after injection. Thereafter, mean serum
(Fig. 15.11; Table 15.2). concentration remained in the normal range up to
Because of the promising results of the first clin- week seven after injection in the 500 mg dose group
ical study with testosterone buciclate, a follow-up and at least up to week nine in the 1000 mg dose
study was initiated. After complete wash-out from group. The terminal elimination half-lives were
previous therapy, all hypogonadal men received a 18.3  2.3 and 23.7  2.7 days for the 500 mg dose
single im injection of 1000 mg testosterone buciclate. and 1000 mg dose groups, respectively. Administra-
As in the previous study with lower doses, no initial tion of 500 mg of this testosterone preparation every
burst release of testosterone was observed. Maximal four weeks, after an initial loading dose of 1000 mg,
testosterone serum levels were observed nine weeks for up to 12 months, to 308 healthy men for male
(tmax) after injection, with a mean value of 13.1 contraception, maintained serum levels of testoster-
 1.8 nmol/l (cmax). Following peak concentrations, one in the normal range when measured directly
testosterone serum levels gradually declined and before the next injection (Gu et al. 2003).
remained within the normal range up to week 16. In the first study in Caucasian men, im injections
This study demonstrated that an increase of the of 250 mg or 1000 mg testosterone undecanoate in tea

321
Chapter 15: Testosterone preparations for clinical use

seed oil were given to 14 hypogonadal patients (Behre six-week intervals. Following the first injection, mean
et al. 1999a). Follow-up examinations were performed serum levels of testosterone were never found below
1, 2, 3, 5 and 7 days after injection and then weekly up the lower limit of normal (Fig. 15.13). However, peak
to study week eight. Whereas no prolonged increase and trough serum concentrations of testosterone
of testosterone was observed in the 250 mg group, increased during the six-month treatment, with tes-
serum levels of testosterone in the higher dose group tosterone levels above the upper normal limit after the
increased from 4.8  0.9 nmol/l (mean  SEM) to third and fourth injection. Therefore, in 7 of the 14
maximum levels of 30.5  4.3 nmol/l at day 7 (tmax). hypogonadal men, injections were given at gradually
Testosterone levels remained within the normal range increasing intervals between the 5th and 10th injec-
up to week seven (13.5  1.2 nmol/l). Non-linear tion, and from then on every 12 weeks (von Eckard-
regression analysis revealed a terminal elimination stein and Nieschlag 2002). During steady state, serum
half-life for intramuscular testosterone undecanoate levels of testosterone remained in the normal range
of 20.9  6.0 days (Fig. 15.12). with maximal concentrations of 32.0  11.7 nmol/l
Similar to the preclinical study in monkeys, the (mean  SD) one week after injection, and nadir
clinical study in hypogonadal men demonstrated levels before the next injection of 12.6  3.7 nmol/l
favorable pharmacokinetics of intramuscular testos- (Fig. 15.14).
terone undecanoate. Because of the relatively low During the last decade, testosterone undecanoate
concentration of 125 mg testosterone undecanoate dissolved in castor oil became one of the favorite intra-
per milliliter of tea seed oil, however, administration muscular testosterone esters for treatment of hypogo-
of the 1000 mg dose requires an injection volume nadal men (see Chapter 14) and for clinical studies of
of 8 ml, which renders im administration impractic-
able. Therefore, the preparation was reformulated
and testosterone undecanoate dissolved in castor 36
33
oil at a higher concentration of 250 mg/ml.
Testosterone (mmol/l)

30
Fourteen hypogonadal patients received one im 27
injection of 1000 mg of the reformulated testoster- 24
21
one undecanoate preparation (Behre et al. 1999a). 18
Maximal serum levels with the reformulated 15
12
preparation were lower than with the Chinese prep- 9
aration and remained within the mid-normal range 6
(Fig. 15.12). Pharmacokinetic analysis revealed a 3
0
long terminal elimination half-life of 33.9  4.9 days 0 7 14 21 28 35 42 49 56
(Table 15.2). Time (d)
Due to these favorable pharmacokinetics, a first, Fig. 15.12 Serum concentrations (mean  SEM) of testosterone
prospective, open-label study with repeated im injec- after single-dose im injections of 1000 mg testosterone
tion was initiated (Nieschlag et al. 1999). Thirteen undecanoate in tea seed oil in 7 hypogonadal men () or castor oil
in 14 hypogonadal men (). Broken lines indicate normal range of
hypogonadal men received four im injections of testosterone (from Behre et al. 1999a, adapted with permission of
1000 mg testosterone undecanoate in castor oil at the European Journal of Endocrinology).

50 Fig. 15.13 Serum concentrations (mean


 SEM) of testosterone after multiple im
Testosterone (nmol/l)

40 injections of 1000 mg testosterone


undecanoate in castor oil in 13
30 hypogonadal men. Broken lines indicate
normal range of testosterone (adapted
20 with permission from Nieschlag et al.
1999, copyright 1999, Blackwell
10 Publishing).
0
2 0 2 4 6 8 10 12 14 16 18 20 22 24
Time (weeks)

322
Chapter 15: Testosterone preparations for clinical use

40 of about 12 weeks, as demonstrated for testosterone


35 undecanoate in hypogonadal men.
Testosterone (nmol/l)

30
25 15.6.8 Testosterone microcapsules
Testosterone can be encapsuled in a biodegradable
20
matrix composed of lactide/glycolide copolymer
15 which is suitable for im injection. When microcapsule
10 injections containing 315 mg of testosterone were
5
given to eight hypogonadal men, serum testosterone
levels slowly increased to peak levels at about 8 weeks
0
0 7 14 21 28 35 42 49 56 63 70 77 84
and fell thereafter to reach pathological levels again by
Time (d)
11 weeks (Burris et al. 1988). In later studies the size-
range and the testosterone loading of the microcap-
Fig. 15.14 Serum concentrations (mean  SD) of testosterone
after single injection of 1000 mg testosterone undecanoate in castor
sules were adjusted so that, in hypogonadal men, im
oil in hypogonadal men () and during multiple injections with the injections of 630 mg of testosterone in microcapsules
same dose every 12 weeks (). Broken lines indicate normal range of (two im injections, 2.5 ml volume for each injection)
testosterone (adapted with permission from von Eckardstein and
Nieschlag 2002).
resulted in serum levels of testosterone within the
normal range for about 70 days (Bhasin et al. 1992).
Similar results were obtained after subcutaneous
hormonal male contraception (see Chapter 22). Large- injection of up to 534 mg of testosterone in micro-
scale studies involving 1493 hypogonadal patients, com- capsules (total injection volume up to 5 ml) in hypo-
prising 6333 im injections and 1103 patient-years of gonadal men, albeit with a pronounced early peak and
treatment, demonstrated that this form of testosterone a relatively long period of low-normal serum total
treatment is well tolerated, effective and safe (Zitzmann testosterone (Amory et al. 2002). These clinical stud-
and Nieschlag 2007; Zitzmann et al. 2011). ies demonstrated that the microspheres can be
adapted to the required needs, but further long-term
studies are needed to evaluate the clinical potential of
15.6.7 Testosterone decanoate this form of testosterone administration.
Testosterone decanoate differs from testosterone
undecanoate by one carbon atom in the ester side 15.7 Testosterone pellets for
chain. It has been widely administered for many
years as part of a mixture with shorter-action testos- subdermal administration
terone esters; however, it has not been available as a Subdermal testosterone pellet implantation was
single preparation. To date there are no detailed among the earliest effective modalities employed for
studies published on the pharmacokinetics of admin- clinical application of testosterone, which became an
istration of testosterone decanoate to hypogonadal established form of androgen replacement therapy by
men. However, im injections of 400 mg of testoster- 1940 (Deanesly and Parkes 1938; Vest and Howard
one decanoate were given four times at injection 1939). With the advent of other modalities, e.g. intra-
intervals of four weeks to normal men in a contra- muscular testosterone ester injections, it went out of
ceptive study (Anderson et al. 2002). Endogenous general use. However, investigations in the 1990s
testosterone was suppressed by concomitant admin- redefined the favorable pharmacokinetic profiles
istration of etonogestrel implants. Nadir testosterone and clinical pharmacology of testosterone implants
levels before the next injection were in the lower (Handelsman et al. 1990; Jockenhvel et al. 1996).
normal range; whereas serum levels were at the The original testosterone implants were manu-
upper normal limit one week after injection. From factured by high-pressure tableting of crystalline
these limited data it can be concluded that testoster- steroid with a cholesterol excipient. These proved
one decanoate seems to have an improved pharma- brittle, hard to standardize or sterilize and exhibited
cokinetic profile over testosterone enanthate, but surface unevenness and fragmentation during in-
does not allow similar prolonged injection intervals vivo absorption, to produce an uneven late release

323
Chapter 15: Testosterone preparations for clinical use

rate. These limitations were overcome in the 1950s In a recent randomized, crossover clinical trial
by switching to high-temperature molding whereby comparing subdermal testosterone pellets versus
molten testosterone was cast into cylindrical moulds injectable testosterone undecanoate in 38 hypogona-
to produce more robust implants. These have more dal patients, no consistent differences were seen in a
uniform composition, resulting in a more steady comprehensive range of pharmacodynamic measures
and prolonged release and reduced tissue reaction. reflecting androgen effects on biochemistry and
Sterilization is achieved by a combination of high- hematology, muscle mass and strength, quality of life,
temperature exposure during manufacture together mood and sexual function (Fennell et al. 2010). How-
with surface sterilization or, more recently, gamma- ever, 91% of men preferred the injectable over the
irradiation. The testosterone implants are currently implantable form of testosterone therapy.
available in two sizes with a common diameter of
4.5 mm: 6 mm length for the 100 mg and 12 mm 15.8 Testosterone preparations for
length for the 200 mg implant. Pellets are usually
implanted under the skin of the lower abdominal transdermal administration
wall under sterile conditions using a trochar and 15.8.1 Testosterone patches
cannula.
The estimated half-life of absorption of testoster- The skin easily absorbs steroids and other drugs, and
one from subdermal implants is 2.5 months. On transdermal drug delivery has become a widely used
average, approximately 1.3 mg of testosterone is therapeutic modality. The scrotum shows the highest
released per day from the 200 mg pellet. Testosterone rate of steroid absorption, about 40-fold higher than
implants demonstrate a minor and transient acceler- the forearm (Feldmann and Maibach 1967). This
ated initial burst release, which lasts for 12 days difference in absorption rates has been exploited for
(Jockenhvel et al. 1996). The most comprehensive the development of a transdermal therapeutic system
pharmacokinetic evaluation of testosterone implants (TTS) to deliver testosterone. Large 40 and 60 cm2
was conducted in a random-sequence, crossover clin- polymeric membranes loaded with 10 or 15 mg tes-
ical study of 43 androgen-deficient men with primary tosterone, when attached to the scrotal skin deliver
or secondary hypogonadism (Handelsman et al. sufficient amounts of the steroid to provide hypogo-
1990). Patients were treated sequentially with three nadal men with serum levels in the physiological
regimens six 100 mg, three 200 mg or six 200 mg range (Bals-Pratsch et al. 1986; 1988; Findlay et al.
implants at intervals of at least six months. Implant- 1987; Korenman et al. 1987). The application of the
ation of testosterone pellets resulted in a highly repro- patch to scrotal skin requires hair clipping or shaving
ducible and dose-dependent time-course for
circulation of total and free testosterone. Testosterone 40
concentrations reached baseline by six months after
either of the 600 mg dose regimens but remained
Testosterone (nmol/l)

30
significantly elevated after six months following the
1200 mg dose. The standard dose for hypogonadal
men is 800 mg every six months, which can be titrated 20
individually (Fig. 15.15).
Pellet implantation is generally well tolerated
(Handelsman et al. 1997; Kaminetsky et al. 2011). 10
Adverse events after implantations were extrusions
(8.512% per procedure), bruising (2.38.8%) and 0
infections (0.64%) (Handelsman et al. 1997; Kelleher 0 1 2 3 4 5 6
et al. 1999). Due to the long-lasting effect and the Time (months)
inconvenience of removal, preferably pellets should Fig. 15.15 Blood total testosterone (mean  SEM) in 43
be used by men in whom the beneficial effects and hypogonadal men receiving four 200 mg pellets (800 mg) implanted
tolerance for androgen replacement therapy have either under the skin of the lateral abdominal wall (in 4 tracks ();
n = 9, or in 2 tracks (); n = 16) or in the hip region (; n = 18).
already been established by treatment with shorter- (Adapted with permission from Kelleher et al. 2001, copyright 2001,
acting testosterone preparations. Blackwell Publishing.)

324
Chapter 15: Testosterone preparations for clinical use

Fig. 15.16 Serum concentrations (mean


35  SEM) of testosterone () and DHT () in
11 hypogonadal men before and during
30 treatment with trans-scrotal testosterone
patches. Broken lines indicate normal
Testosterone (nmol/l)

range of testosterone, dotted line upper


25 normal limit of DHT (adapted with
permission from Behre et al. 1999b,
DHT (nmol/l)

20 copyright 1999, Blackwell Publishing).

15

10

0
0 1 2 3 4 5 6 7 8 9 10
Time (years)

to optimize adherence. The membranes need to be severity of skin irritation, although the effects on
renewed every day. When applied in the morning and pharmacokinetics of testosterone are unclear.
worn until the next morning the resulting serum Recently, a new thin, transparent and comfortable
testosterone levels resemble the normal diurnal vari- testosterone-in-adhesive matrix patch was launched
ations of serum testosterone in normal men without that has to be applied only every second day. The
supraphysiological peaks (Bals-Pratsch et al. 1988). adhesiveness seems to be quite good, even after phys-
Long-term therapy up to 10 years with daily adminis- ical exercise and after sauna (Raynaud et al. 2009).
tration of the scrotal patch in 11 hypogonadal men Depending on the size of the patch (30, 45 or 60 cm2),
produced steady-state serum levels of testosterone the average daily release is 1.2, 1.8 or 2.4 mg of
and estradiol in the normal range, and serum levels testosterone. The recommended initial dosing regi-
of DHT at or slightly above the higher limit of normal men is two 60 cm2 patches every two days, which
without significant adverse side-effects (Fig. 15.16; can be modified depending on the serum levels of
Behre et al. 1999b). However, with the introduction testosterone achieved (Fig. 15.18). In a randomized,
of testosterone gels for treatment of hypogonadism, open label, multicenter, one-year study involving 224
scrotal testosterone patches lost popularity and the hypogonadal patients, testosterone serum levels were
marketing has been discontinued. above 3 ng/ml in 85% of patients after application of
While testosterone is readily absorbed by genital two patches of 60 cm2 every 48 h, and remained stable
skin, transdermal systems for use on non-genital skin over 12 months of testosterone therapy (Raynaud
require enhancers to facilitate sufficient testosterone et al. 2008b).
passage through the skin. The permeation-enhanced
testosterone patch delivers 5 mg/day testosterone
when applied to non-scrotal skin. If one or two such 15.8.2 Testosterone gels
systems are worn for 24 hours physiological serum In 2000, a 1% colorless hydroalcoholic gel containing
testosterone levels can be mimicked, as with scrotal 25 or 50 mg testosterone in 2.5 or 5 g gel was
patches (Fig. 15.17) (Brocks et al. 1996; Meikle et al. approved for clinical use in hypogonadism. The gel
1996). Due to the alcoholic enhancer used and the dries in less than 5 min without leaving a visible
occlusive nature of the systems, the application is residue on the skin. About 9 to 14% of the testoster-
associated with skin irritation in up to 60% of the one in the gel is bioavailable. Application of the tes-
subjects, with most users discontinuing application tosterone gel increased serum testosterone levels into
because of the skin irritation (Jordan 1997; Parker the normal range within one hour after application
and Armitage 1999). Preapplication of corticosteroid (Wang et al. 2000). Steady-state serum levels are
cream to the skin has been reported to decrease the achieved 4872 hours after initiation of therapy;

325
Chapter 15: Testosterone preparations for clinical use

42 1200 Fig. 15.17 Serum concentrations (mean


 SD) of testosterone during and after
nighttime application of two non-scrotal
testosterone systems to the backs of 34
35 1000 hypogonadal men. Shaded area indicates
normal range of testosterone (adapted
with permission from Meikle et al. 1996,
copyright 1996, The Endocrine Society).
Testosterone (nmol / l)

28 800

(ng / dl)
21 600

14 400

7 200

0 0
0 12 24 36 48

Time (h)

8.00 Fig. 15.18 Serum concentrations (mean


 SEM) of testosterone during single 48 h
7.00 application of two matrix patches (, 30
cm2; , 45 cm2; , 60 cm2) and 13 h
6.00 after removal (adapted with permission
from Raynaud et al. 2008a).
Testosterone (ng/ml)

5.00

4.00

3.00

2.00

1.00

0.00
0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 54
Time (h)

whereas pretreatment serum testosterone levels are Long-term pharmacokinetics of the transdermal
seen four days after stopping application. The appli- testosterone gel were evaluated in 227 hypogonadal
cation of the testosterone gel at four sites (application men (Swerdloff et al. 2000). Patients were randomly
skin areas approximately four times that of one site) assigned to application of 5 or 10 g of the testosterone
resulted in an AUC of testosterone which was 23% gel or two patches of a non-scrotal testosterone
higher compared to application of the same amount system. After 90 days of testosterone gel treatment,
of gel on one site. However, this difference did not the dose was titrated up (5 to 7.5 g) or down (10 to 7.5 g)
achieve statistical significance in the nine hypogona- if the preapplication serum testosterone levels were
dal men tested (Wang et al. 2000). outside the normal adult male range. During

326
Chapter 15: Testosterone preparations for clinical use

Day 0 Day 1 Day 30


40
40 40

30
30 30

20 20
20

10 10 10
Serum testosterone (nmol/l)

0 0 0
0 8 16 24 0 8 16 24 0 8 16 24

Day 90 Day 180


40 40 T-Patch
T-Gel 100 mg
30 30 T-Gel 50 mg
T-Gel 50 75 mg
20 T-Gel 100 75 mg
20

10 10

0 0
0 8 16 24 0 8 16 24
Time (h)
Fig. 15.19 Serum concentrations (mean  SEM) of testosterone before (day 0) and after transdermal testosterone applications on days 1, 30, 90
and 180. Time 0 was 8 a.m., when blood sampling usually began. On day 90, the dose in the subjects applying testosterone gel (T gel) 50 or 100
mg was up- or down-titrated if their preapplicaton serum testosterone levels were below or above the normal adult male range, respectively.
Dotted lines denote the adult normal range (adapted with permission from Swerdloff et al. 2000, copyright 2000, The Endocrine Society).

long-term treatment mean serum levels of testoster- Since then, a number of other testosterone gels
one were maintained in the mid-normal range with have been under development. Two randomized,
5 g of gel and in the upper normal range with 10 g controlled studies demonstrated a dose-dependent
of gel (Fig. 15.19). Testosterone gel application increase of testosterone serum levels to the normal
resulted in dose-proportionate increases in serum range in hypogonadal men after 90 days of applica-
DHT and estradiol as well as dose-proportionate tion of 5 g/d or 10 g/d of another hydroalcoholic
decreases of gonadotropins. topical gel containing 1% testosterone compared to
The advantages of the testosterone gel over non-scrotal testosterone patches (n = 208; McNicholas
the early testosterone patches are a lower incidence et al. 2003) or compared to non-scrotal testosterone
of skin irritation, the ease of application, the invisi- patches and placebo gel (n = 406; Steidle et al. 2003).
bility of the dried gel, and the ability to deliver testos- Recent data from a registry study in 849 hypogonadal
terone dose dependently to the low, mid- or upper patients treated over 12 months demonstrated high
normal range. A potential adverse side-effect of compliance with this form of testosterone replacement
testosterone gel application is the transfer of testoster- (Khera et al. 2011).
one to women or children upon close contact with A new testosterone gel at a concentration of 2%
the skin. Transfer of transdermal testosterone from instead of the 1% of the earlier preparations
the skin can be avoided by applying gel to skin was launched first in Sweden and Germany in 2005.
covered by clothing, or showering after application. This gel is provided in a metered-dose pump allowing
This preparation has gained a significant market adjusted dosing starting from 10 mg testosterone
share of androgen formulations in Europe and the (0.5 g, 1 pump actuation) up to higher doses in
United States. 10-mg steps. The recommended starting dose is

327
Chapter 15: Testosterone preparations for clinical use

4060 mg of testosterone, e.g. 46 pump actuations or effect on normalization of testosterone concentration


23 g of the testosterone gel per day. In a recent large- after application of this testosterone gel in hypogona-
scale, placebo-controlled study involving 220 hypogo- dal men, and at least 75% of patients achieved serum
nadal men with diabetes mellitus and/or metabolic levels in the normal range after three months of gel
syndrome, good safety profile of this testosterone application (Dobs et al. 2011).
preparation was demonstrated, with no significant To date, the testosterone gel with the highest con-
differences in the frequencies of adverse events or centration of testosterone is a 2.5% topical gel.
serious adverse events between patients treated with Recently, it has been launched in sachets containing
testosterone or placebo gel (Jones et al. 2011). Hypo- 2.5 g gel (62.5 mg testosterone) or 5 g gel (125 mg
gonadal patients applied a starting dose of 3 g of the testosterone). Application of 5 g/d of this 2.5% hydro-
2% testosterone gel (60 mg testosterone), and dose alcoholic gel increased serum levels of testosterone to
adjustments to 2 g (40 mg testosterone) or 4 g (80 mg the normal range in 14 gonadotropin-suppressed
testosterone) were made dependent on the serum normal men (Rolf et al. 2002a). Washing of the skin
levels of testosterone measured. In another recent after 10 minutes did not influence the pharmacoki-
study it was demonstrated that BMI has no significant netic profile. No interpersonal testosterone transfer

(A)
50

40

30
Testosterone or testosterone + DHT (nmol/l)

20



10

0
(B)
50

40

30


20

10

0

0 2 4 8 12 16 20 24
Time (weeks)
Fig. 15.20 Median and box plots of testosterone serum concentrations (A) and testosterone plus DHT serum concentrations (B), measured at
10 a.m., at weeks 024 in patients treated with the dermal 2.5% testosterone gel (), scrotal 2.5% testosterone gel () and non-scrotal
testosterone patch () (from Khnert et al. 2005, adapted with permission of the European Journal of Endocrinology).

328
Chapter 15: Testosterone preparations for clinical use

could be detected after evaporation of the alcohol serum levels of testosterone (with dose adjustments
vehicle of this testosterone gel (Rolf et al. 2002b). In a to 1 g of 1% testosterone gel to 2.5 g of 2.5% testoster-
randomized, controlled, multicenter clinical trial, ini- one gel). Unfortunately, the scrotal application, despite
tial application of 5 g of the testosterone gel, with the being well tolerated by the patients, has not been
possibility of dose adjustment down to 2.5 g or up to approved by regulatory bodies.
10 g after four and eight weeks of treatment, resulted in Recently, the initial 1% hydroalcoholic testosterone
significantly higher testosterone serum levels, well gel has been reformulated to a 1.62% gel with increased
within the normal range, compared to a control group viscosity and increased skin permeation, allowing deliv-
treated with non-scrotal testosterone patches (initially ery of testosterone also with a lower volume of gel. In a
2 patches, with individual adjustments to 13 patches multicenter, randomized, placebo-controlled study, 2.5 g
per day) (Fig. 15.20; Khnert et al. 2005). In the same of this gel preparation was applied once daily for up to
study it could be demonstrated that scrotal application 182 days to either upper arms/shoulders or abdomen of
of only 1 g of the testosterone gel also normalized hypogonadal patients (Kaufman et al. 2011; Fig. 15.21).

Fig. 15.21 Mean concentrations of serum testosterone on day 14, 56, 112 and 182 after daily application of 1.62% dermal testosterone gel.
Dotted lines indicate normal testosterone range. On day 14, all patients in the testosterone group applied 2.5 g gel (adapted with permission
from Kaufman et al. 2011).

329
Chapter 15: Testosterone preparations for clinical use

Fig. 15.22 Serum concentrations (mean  SD)


of testosterone at day 15, 60 and 120 in
hypogonadal patients after daily application of
testosterone topical solution to the axillae
(adapted with permission from Wang et al. 2011).

Dose adjustments down to 1.25 g and up to 3.75 g or 5.0 g using an applicator, so it might be a favorable new
were made 14 d, 28 d or 42 d after initiation of treatment option for testosterone replacement of hypogonadal
depending on serum testosterone levels. After titration, men.
7.3% of patients were treated with 1.25 g/d, 25.6% with
2.5 g/d, 28.2% with 3.75 g/d and 38.9% with 5 g/d. After 15.9 Key messages
182 days of therapy, 82.2% of hypogonadal patients  Oral, buccal, injectable, subdermal implantable and
achieved average serum levels of testosterone within transdermal testosterone preparations are
the normal eugonadal range. available for clinical use. The best preparation is the
one that replaces testosterone serum levels at as
close to physiological concentrations as possible.
15.8.3 Testosterone topical solution  Oral administration of testosterone undecanoate
A 2% formulation of a testosterone solution was in oleic acid or castor oil results in high
approved by the FDA in November 2010. It is a interindividual and intraindividual variability of
non-occlusive topical formulation administered to serum testosterone values. A new oral
the axilla, with an applicator instead of with the hands testosterone undecanoate preparation (proprietary
self-emulsifying drug delivery system) shows an
as for the testosterone gels. The safety, pharmacoki-
improved pharmacokinetic profile.
netics and efficacy of the topical solution were evalu-
 Daily or twice-daily buccal administration of
ated in an open-label, multicenter clinical trial (Wang testosterone tablets increases serum testosterone
et al. 2011). All hypogonadal patients started with a to the normal range. Relatively low acceptability of
daily dose of 60 mg of the testosterone solution. Doses this application form seems to be a disadvantage.
could be adjusted on days 45 and 90 to achieve serum  The older testosterone esters for im injection
levels of testosterone in the normal range. Most of the (testosterone propionate, testosterone
patients (97/135) stayed at the 60 mg dose after 120 enanthate, testosterone cypionate, testosterone
days of therapy; 3/135 used the 30 mg dose; 25/135 cyclohexanecarboxylate) are still widely used, but
the 90 mg dose; and 10/135 the 120 mg dose. After are suboptimal for the treatment of male
four months of therapy, 84.1% of hypogonadal hypogonadism. Doses and injection intervals
most frequently used in the clinic lead to initial
patients had average serum levels of testosterone
supraphysiological testosterone levels and
within the normal range (10.436.4 nmol/l) (Fig. subnormal values before the next injection. To
15.22). Application site erythema was noted in 5.2% obtain testosterone serum concentrations
of patients. This mode of testosterone application continuously in the normal range, unacceptably
might reduce the undesired transfer of testosterone frequent small doses would have to be injected.
to other persons, as it is applied not by hand but by

330
Chapter 15: Testosterone preparations for clinical use

 Intramuscular injection of 1000 mg  Daily administration of testosterone gel increases


testosterone undecanoate to hypogonadal men serum levels of testosterone in hypogonadal
maintains serum levels of testosterone within the patients dose-dependently to the normal range.
normal range for three months. Injection Whereas skin irritation is minimal, a potential
intervals can be adjusted for individual patient adverse side-effect of testosterone gel
needs. application is the transfer of testosterone to
 A single implantation procedure of testosterone women or children upon close contact with
pellets provides serum levels of testosterone in the skin. Various gel preparations have been
the normal range for up to six months. Pellet launched with testosterone concentrations
extrusion occurs in about 10% of the of 12.5%.
implantation procedures. Due to the long-lasting  If a 2.5% testosterone gel is applied to the scrotal
effect and the inconvenience of removal, skin, only about 20% of the amount of gel
preferably pellets should be used by men in needed for non-genital application is required for
whom the beneficial effects and tolerance for substitution.
androgen replacement therapy have already  Administration of a 2% formulation of a
been established. topical testosterone solution applied to
 Transdermal application of testosterone by the axilla using a special applicator
scrotal or non-scrotal patches increases serum normalizes serum testosterone levels in 84%
levels of testosterone to the normal range and of hypogonadal men after four months
even mimics the physiological circadian of treatment and has a good skin
testosterone rhythm. safety profile.

15.10 References enanthate-induced azoospermia and


oligozoospermia in a male
Bals-Pratsch M, Langer K, Place VA,
Nieschlag E (1988) Substitution
Aakvaag A, Stromme SB (1974) The contraceptive study. II. therapy of hypogonadal men with
effect of mesterolone administration Pharmacokinetics and transdermal testosterone over one
to normal men on the pituitary- pharmacodynamics of once weekly year. Acta Endocrinol (Copenh)
testicular function. Acta Endocrinol administration of testosterone 118:713
77:380386 enanthate. J Clin Endocrinol Metab Behre HM, Nieschlag E (1992)
Alkalay D, Khemani L, Wagner WE, 81:896901 Testosterone buciclate (20 Aet-1) in
Bartlett MF (1973) Sublingual and
Anderson RA, Zhu H, Cheng L, Baird hypogonadal men: pharmacokinetics
oral administration of
DT (2002) Investigation of a novel and pharmacodynamics of the new
methyltestosterone. A comparison
preparation of testosterone long-acting androgen ester. J Clin
of drug bioavailability. J Clin
decanoate in men: Endocrinol Metab 75:12041210
Pharmacol New Drugs
pharmacokinetics and
13:142151 Behre HM, Baus S, Kliesch S, Keck C,
spermatogenic suppression Simoni M, Nieschlag E (1995)
Amory JK, Anawalt BD, Blaskovich with etonogestrel
PD, Gilchriest J, Nuwayser ES, Potential of testosterone buciclate
implants. Contraception for male contraception: endocrine
Matsumoto AM (2002) 66:357364
Testosterone release from a differences between responders and
subcutaneous, biodegradable Anonymous (1988) Methyltestosteron- nonresponders. J Clin Endocrinol
microcapsule formulation (Viatrel) Monographie. Bundesanzeiger Metab 80:23942403
in hypogonadal men. J Androl 40:51405141 Behre HM, Abshagen K, Oettel M,
23:8491 Bagchus WM, Hust R, Maris F, Hubler D, Nieschlag E (1999a)
Amory JK, Bush MA, Zhi H, Caricofe Schnabel PG, Houwing NS (2003) Intramuscular injection of
RB, Matsumoto AM, Swerdloff RS, Important effect of food on the testosterone undecanoate for the
Wang C, Clark RV (2011) Oral bioavailability of oral testosterone treatment of male hypogonadism:
testosterone with and without undecanoate. Pharmacotherapy phase I studies. Eur J Endocrinol
concomitant inhibition of 23:319325 140:414419
5a-reductase by dutasteride in Bals-Pratsch M, Knuth UA, Yoon YD, Behre HM, von Eckardstein S,
hypogonadal men for 28 days. Nieschlag E (1986) Transdermal Kliesch S, Nieschlag E (1999b)
J Urol 185:626632 testosterone substitution therapy Long-term substitution therapy of
Anderson RA, Wu FC (1996) for male hypogonadism. Lancet hypogonadal men with
Comparison between testosterone 2:943946 transscrotal testosterone over

331
Chapter 15: Testosterone preparations for clinical use

710 years. Clin Endocrinol (Oxf) Cunningham GR, Silverman VE, cross-over clinical trial of injectable
50:629635 Kohler PO (1978) Clinical vs. implantable depot testosterone
Bhasin S, Swerdloff RS, Steiner B, evaluation of testosterone enanthate for maintenance of testosterone
Peterson MA, Meridores T, for induction and maintenance of replacement therapy in androgen
Galmirini M, Pandian MR, reversible azoospermia in man. In: deficient men. Clin Endocrinol (Oxf)
Goldberg R, Berman N (1992) Patanelli DJ (ed) Hormonal Control 73:102109
A biodegradable testosterone of Male Fertility (Workshop Findlay JC, Place VA, Snyder PJ (1987)
microcapsule formulation provides Proceedings), NIH Publication No. Transdermal delivery of
uniform eugonadal levels of 781097. US Department of Health, testosterone. J Clin Endocrinol
testosterone for 1011 weeks in Education, and Welfare (NIH), Metab 64:266268
hypogonadal men. J Clin Endocrinol Bethesda, MD, pp 7187
Frey H, Aakvaag A, Saanum D, Falch J
Metab 74:7583 Danner C, Frick G (1980) Androgen (1979) Bioavailability of oral
substitution with testosterone- testosterone in males. Eur J Clin
Boyd PR, Mark GJ (1977) Multiple
containing nasal drops. Int J Androl Pharmacol 16:345349
hepatic adenomas and a
3:429431
heptatocellular carcinoma in a man Fujioka M, Shinohara Y, Baba S, Irie
on oral methyl testosterone for Deanesly R, Parkes AS (1938) Further M, Inoue K (1986) Pharmacokinetic
eleven years. Cancer 40: experiments on the administration properties of testosterone
17651770 of hormones by the subcutaneous propionate in normal men.
implantation of tablets. Lancet J Clin Endocrinol Metab
Breuer H, Gtgemann D (1966) 232:606608
Wirkung von 1alpha-Methyl- 63:13611364
5alpha-androstan-17b-ol-3-on de Lorimer AA, Gordan GS, Lwe RC, Fukutani K, Isurugi K, Takayasu H,
(Mesterolon) auf die Carbone JV (1965) Wakabayashi K, Tamaoki B-I
Steroidausscheidung beim Methyltestosterone: related steroids (1974) Effects of depot testosterone
Menschen. Arzneimittelforschung and liver function. Arch Intern Med therapy on serum levels of
16:759762 116:289294 luteinizing hormone and follicle-
Brocks DR, Meikle AW, Boike SC, Dobs A, McGettigan J, Norwood P, stimulating hormone in patients
Mazer NA, Zariffa N, Audet PR, Potts S, Gould E (2011) Baseline with Klinefelters syndrome and
Jorkasky DK (1996) body mass index (BMI) has no hypogonadotropic eunuchoidism.
Pharmacokinetics of testosterone effect upon normalization of J Clin Endocrinol Metab 39:856864
in hypogonadal men after testosterone concentrations with Gerhards E, Gibian H, Kolb KH (1966)
transdermal delivery: influence of testosterone 2% gel [abstract]. Zum Stoffwechsel von 1alpha-
dose. J Clin Pharmacol 36:732739 Endocr Rev 32 Methyl-5alpha androstan-17b-ol-3-
(03_MeetingAbstracts):P1353 on (Mesterolon) beim Menschen.
Burris AS, Ewing LL, Sherins RJ (1988)
Escamilla RF (1949) Treatment of Arneimittelforschung 16:458463
Initial trial of slow-release
testosterone microspheres in preadolescent eunuchoidism with Goodman MA, Laden AMJ (1977)
hypogonadal men. Fertil Steril (methyl)testosterone linguets. Amer Hepatocellular carcinoma in
50:493497 Pract 3:425 association with androgen therapy.
Falk H, Thomas LB, Popper H, Med J Aust 1:220221
Carbone JV, Grodsky GM, Hjelte V
(1959) Effect of hepatic dysfunction Ishak KG (1979) Hepatic Gordon RD, Thomas MJ, Poyntin JM,
on circulating levels of angiosarcoma associated with Stocks AE (1975) Effect of
sulphobromopthalein and androgenic-anabolic steroids. mesterolone on plasma LH, FSH and
its metabolites. J Clin Invest Lancet 2:11201123 testosterone. Andrologia 7:287296
38:19891996 Farrell GC, Joshua DE, Uren RF, Gu YQ, Wang XH, Xu D, Peng L,
Coert A, Geelen J, de Visser J, van der Baird PJ, Perkins KW, Cheng LF, Huang MK, Huang ZJ,
Vies J (1975) The pharmacology Kronenberg H (1975) Zhang GY (2003) A multicenter
and metabolism of testosterone Androgen-induced hepatoma. contraceptive efficacy study of
undecanoate (TU), a new orally Lancet 1:430432 injectable testosterone
active androgen. Acta Endocrinol Feldmann RJ, Maibach HI (1967) undecanoate in healthy Chinese
79:789800 Regional variation in percutaneous men. J Clin Endocrinol Metab
penetration of 14C cortisol in man. 88:562568
Crabb P, Diczfalusy E, Djerassi C
(1980) Injectable contraceptive J Invest Dermatol 48:181183 Hamburger C (1958) Testosterone
synthesis: an example of Fennell C, Sartorius G, Ly LP, treatment and 17-ketosteroid
international cooperation. Science Turner L, Liu PY, Conway AJ, excretion. Acta Endocrinol
209:992994 Handelsman DJ (2010) Randomized 28:529536

332
Chapter 15: Testosterone preparations for clinical use

Handelsman DJ, Conway AJ, Boylan comparison of scrotal and nonscrotal Khnert B, Byrne M, Simoni M,
LM (1990) Pharmacokinetics and transdermal systems. Am J Contact Kpcke W, Gerss J, Lemmnitz G,
pharmacodynamics of testosterone Dermat 8:108113 Nieschlag E (2005) Testosterone
pellets in man. J Clin Endocrinol Junkmann K (1952) ber protrahiert substitution with a new
Metab 70:216222 wirksame Androgene. Arch Path transdermal, hydroalcoholic gel
Pharmacol 215:8592 applied to scrotal or non-scrotal
Handelsman DJ, Mackey MA, Howe C,
skin: a multicentre trial. Eur
Turner L, Conway AJ (1997) Junkmann K (1957) Long-acting J Endocrinol 153:317326
Analysis of testosterone implants steroids in reproduction. Recent
for androgen replacement therapy. Prog Horm Res 13:389419 Lee A, Rubinow K, Clark RV, Caricofe
Clin Endocrinol 47:311316 RB, Bush MA, Zhi H, Roth MY,
Kaminetsky JC, Moclair B, Hemani M, Page ST, Bremner WJ, Amory JK
Heywood R, Hunter B, Green OP, Sand M (2011) A phase IV
Kennedy SJ (1977a) The toxicity of (2011) Pharmacokinetics of
prospective evaluation of the safety modified slow-release oral
methyl testosterone in the rat. and efficacy of extended release
Toxicol Lett 1:2731 testosterone over nine days in
testosterone pellets for the normal men with experimental
Heywood R, Chesterman H, Ball SA, treatment of male hypogonadism. hypogonadism. J Androl Aug 25
Wadsworth PF (1977b) Toxicity of J Sex Med 8:11861196 [Epub ahead of print]
methyl testosterone in the beagle Kaufman JM, Miller MG, Garwin JL, Mackey MA, Conway AJ, Handelsman
dog. Toxicology 7:357365 Fitzpatrick S, McWhirter C, DJ (1995) Tolerability of
Horst HJ, Hltge WJ, Dennis M, Coert Brennan JJ (2011) Efficacy and intramuscular injections of
A, Geelen J, Voigt KD (1976) safety study of 1.62% testosterone testosterone ester in oil vehicle.
Lymphatic absorption and gel for the treatment of Hum Reprod 10:862865
metabolism of orally administered hypogonadal men. J Sex Med
8:20792089 Maisey NM, Bingham J, Marks V,
testosterone undecanoate in man.
English J, Chakraborty J (1981)
Klin Wschr 54:875879 Kelleher S, Conway AJ, Clinical efficacy of testosterone
Jockenhvel F, Vogel E, Kreutzer E, Handelsman DJ (1999) undecanoate in male hypogonadism.
Reinhard W, Lederbogen S, A randomized controlled clinical Clin Endocrinol 14:625629
Reinwein D (1996) trial of antibiotic impregnation of
testosterone pellet implants to Mattern C, Hoffmann C, Morley JE,
Pharmacokinetics and
reduce extrusion rate. Eur Badiu C (2008) Testosterone
pharmacodynamics of
J Endocrinol 146:513518 supplementation for hypogonadal
subcutaneous testosterone implants
men by the nasal route. Aging Male
in hypogonadal man. Clin Kelleher S, Conway AJ, Handelsman DJ 11:171178
Endocrinol 45:6171 (2001) Influence of implantation site
Johnsen SG, Bennet EP, Jensen VG and track geometry on the extrusion McNicholas TA, Dean JD, Mulder H,
(1974) Therapeutic effectiveness rate and pharmacology of Carnegie C, Jones NA (2003)
of oral testosterone. Lancet 2: testosterone implants. Clin A novel testosterone gel
14731475 Endocrinol (Oxf) 55:531536 formulation normalizes androgen
levels in hypogonadal men, with
Johnsen SG, Kampmann JP, Bennet EP, Khera M, Bhattacharya RK, Blick G, improvements in body composition
Jrgensen F (1976) Enzyme Kushner H, Nguyen D, Miner MM and sexual function. BJU Int
induction by oral testosterone. Clin (2011) Improved sexual function 91:6974
Pharmacol Ther 20:233237 with testosterone replacement
therapy in hypogonadal men: real- Meikle AW, Arver S, Dobs AS, Sanders
Jones TH, Arver S, Behre HM, Buvat J, SW, Rajaram L, Mazer N (1996)
Meuleman E, Moncada I, Morales world data from the Testim Registry
in the United States (TRiUS). J Sex Pharmacokinetics and metabolism
AM, Volterrani M, Yellowlees A, of a permeation-enhanced
Howell JD, Channer KS; TIMES2 Med 8:32043213
testosterone transdermal system in
Investigators (2011) Testosterone Korenman SG, Viosca S, Garza D, hypogonadal men: influence of
replacement in hypogonadal men Guralnik M, Place V, Campbell P, application site a clinical research
with type 2 diabetes and/or Davis SS (1987) Androgen therapy center study. J Clin Endocrinol
metabolic syndrome (the of hypogonadal men with Metab 81:18321840
TIMES2 study). Diabetes Care transscrotal testosterone system.
34:828837 Am J Med 83:471478 Minto CF, Howe C, Wishart S, Conway
AJ, Handelsman DJ (1997).
Jordan WP (1997) Allergy and topical Krskemper HI, Noell G (1967) Pharmacokinetics and
irritation associated with transdermal Steroidstruktur und Lebertoxizitt. pharmacodynamics of nandrolone
testosterone administration: a Acta Endocrinol 54:7380 esters in oil vehicle: effects of ester,

333
Chapter 15: Testosterone preparations for clinical use

injection site and injection volume. Raynaud JP, Aumonier C, Gualano V, administration of Andriol
J Pharmacol Exp Ther 281:93102 Betea D, Beckers A (2008a) Testocaps. Clin Endocrinol (Oxf)
Nankin HR (1987) Hormone kinetics Pharmacokinetic study of a new 66:579585
after intramuscular testosterone testosterone-in-adhesive matrix Schulte-Beerbhl M, Nieschlag E
cypionate. Fertil Steril 47:10041009 patch applied every 2 days to (1980) Comparison of testosterone,
hypogonadal men. J Steroid Biochem dihydrotestosterone, luteinizing
Nieschlag E (1981) Ist die Anwendung
Mol Biol 109:177184 hormone, and follicle-stimulating
von Methyltestosteron obsolet?
Dtsch med Wschr 106:11231125 Raynaud JP, Legros JJ, Rollet J, Augs hormone in serum after injection of
M, Bunouf P, Sournac M, Fiet J testosterone enanthate or
Nieschlag E, Mauss J, Coert A,
(2008b) Efficacy and safety of a new testosterone cypionate. Fertil Steril
Kicovic P (1975) Plasma
testosterone-in-adhesive matrix 33:201203
androgen levels in men after oral
patch applied every 2 days for 1 year Schrmeyer T, Nieschlag E (1984)
administration of testosterone or
to hypogonadal men. J Steroid Comparative pharmacokinetics of
testosterone undecanoate. Acta
Biochem Mol Biol 109:168176
Endocrinol 79:366374 testosterone enanthate and
Nieschlag E, Cppers HJ, Raynaud JP, Augs M, Liorzou L, testosterone cyclohexanecarboxylate
Wiegelmann W, Wickings EJ (1976) Turlier V, Lauze C (2009) as assessed by serum and salivary
Adhesiveness of a new testosterone- testosterone levels in normal men.
Bioavailability and LH-suppressing
effect of different testosterone in-adhesive matrix patch after Int J Androl 7:181187
preparations in normal and extreme conditions. Int J Pharm Schrmeyer T, Wickings EJ, Freischem
hypogonadal men. Horm Res 375:2832 CW, Nieschlag E (1983) Saliva and
7:138145 Rolf C, Kemper S, Lemmnitz G, serum testosterone following oral
Eickenberg U, Nieschlag E (2002a) testosterone undecanoate
Nieschlag E, Cppers EJ, Wickings EJ
(1977) Influence of sex, testicular Pharmacokinetics of a new administration in normal and
transdermal testosterone gel in hypogonadal men. Acta Endocrinol
development and liver function on
the bioavailability of oral gonadotrophin-suppressed normal 102:456462
testosterone. Europ J Clin Invest men. Eur J Endocrinol 146:673679 Shackleford DM, Faassen WA,
7:145147 Rolf C, Knie U, Lemmnitz G, Nieschlag Houwing N, Lass H, Edwards GA,
Nieschlag E, Bchter D, von E (2002b) Interpersonal Porter CJ, Charman WN (2003)
Eckardstein S, Abshagen K, Simoni testosterone transfer after topical Contribution of lymphatically
M, Behre HM (1999) Repeated application of a newly developed transported testosterone
intramuscular injections of testosterone gel preparation. Clin undecanoate to the systemic
testosterone undecanoate for Endocrinol (Oxf) 56:637641 exposure of testosterone after oral
substitution therapy in hypogonadal Ruzicka L, Goldberg MW, Rosenberg administration of two Andriol
men. Clin Endocrinol (Oxf) HR (1935) Herstellung des 17- formulations in conscious lymph
51:757763 Methyl-testosterons und anderer duct-cannulated dogs. J Pharmacol
Androsten- und Androstanderivate. Exp Ther 306:925933
Paradinas FJ, Bull TB, Westaby D,
Murray-Lyon IM (1977) Zusammenhnge zwischen Snyder CN, Clark RV, Caricofe RB,
Hyperplasia and prolapse of chemischer Konstitution und Bush MA, Roth MY, Page ST,
hepatocytes into hepatic veins mnnlicher Hormonwirkung. Helv Bremner WJ, Amory JK (2010)
during longterm methyltestosterone Chim Acta 18:14871498 Pharmacokinetics of 2 novel
therapy: possible relationships of Salehian B, Wang C, Alexander G, formulations of modified-release
these changes to the development of Davidson T, McDonald V, Berman oral testosterone alone and
peliosis hepatis and liver tumours. N, Dudley RE, Ziel F, Swerdloff RS with finasteride in normal men
Histopathology 1:225246 (1995) Pharmacokinetics, with experimental
bioefficacy, and safety of sublingual hypogonadism. J Androl 31:
Parker S, Armitage M (1999) 527535
Experience with transdermal testosterone cyclodextrin in
testosterone replacement therapy hypogonadal men: comparison to Snyder PJ, Lawrence DA (1980)
for hypogonadal men. Clin testosterone enanthate a clinical Treatment of male hypogonadism
Endocrinol (Oxf) 50:5762 research center study. J Clin with testosterone enanthate. J Clin
Endocrinol Metab 80:35673575 Endocrinol Metab 51:13351339
Pitha J, Harman SM, Michel ME (1986)
Schnabel PG, Bagchus W, Lass H, Sokol RZ, Swerdloff RS (1986) Practical
Hydrophilic cyclodextrin
derivatives enable effective oral Thomsen T, Geurts TB (2007) The considerations in the use of
effect of food composition on serum androgen therapy. In: Santen JR,
administration of steroidal
hormones. J Pharm Sci 75:165167 testosterone levels after oral Swerdloff RS (eds) Male

334
Chapter 15: Testosterone preparations for clinical use

Reproductive Dysfunction. Marcel Wang C, Eyre R, Clark D, Kleinberg C, methyltestosterone. Lancet


Dekker, New York, pp 211225 Newman I, Iranmanesh A, 2:261263
Steidle C, Schwartz S, Jacoby K, Sebree Veldhuis R, Dudley RE, Berman N, World Health Organization,
T, Smith T, Bachand R; North Davidson T, Barstow TS, Sinow R, Nieschlag E, Wang C, Handelsman
American AA2500 T Gel Study Alexander G, Swerdloff R (1996) DJ, Swerdloff RS, Wu F,
Group (2003) AA2500 testosterone Sublingual testosterone replacement Einer-Jensen N, Waites G (1992)
gel normalizes androgen levels in improves muscle mass and strength, Guidelines for the Use of Androgens.
aging males with improvements in decreases bone resorption and WHO, Geneva
body composition and sexual increases bone formation markers
Wu FCW, Farley TMM, Peregoudov A,
function. J Clin Endocrinol Metab in hypogonadal men a clinical
Waites GMH (1996) Effects of
88:26732681 research center study.
testosterone enanthate in normal
J Clin Endocrinol Metab
men: experience from a multicenter
Stuenkel CA, Dudley RE, Yen SC 81:36543662
(1991) Sublingual administration of contraceptive efficacy study. World
Wang C, Berman N, Longstreth JA, Health Organization Task Force on
testosterone hydroxypropyl-b-
Chuapoco B, Hull L, Steiner B, Methods for the Regulation of Male
cyclodextrin inclusion complex
simulates episodic androgen
Faulkner S, Dudley RE, Swerdloff Fertility. Fertil Steril 65:626636
RS (2000) Pharmacokinetics of
release in hypogonadal men. Yin AY, Htun M, Swerdloff RS, Diaz-
transdermal testosterone gel in
J Clin Endocrinol Metab Arjonilla M, Dudley RE, Faulkner S,
hypogonadal men: application of gel
72:10541059 Bross R, Leung A, Baravarian S,
at one site versus four sites: a
Swerdloff RS, Wang C, Cunningham Hull L, Longstreth JA, Kulback S,
general clinical research center
G, Dobs A, Iranmanesh A, Flippo G, Wang C (2012) Re-
study. J Clin Endocrinol Metab
examination of pharmacokinetics of
Matsumoto AM, Snyder PJ, Weber 85:964969
T, Longstreth J, Berman N (2000) oral testosterone undecanoate in
Wang C, Swerdloff R, Kipnes M, hypogonadal men with a new self-
Long-term pharmacokinetics of
Matsumoto AM, Dobs AS, emulsifying formulation. J Androl
transdermal testosterone gel in
hypogonadal men. J Clin Endocrinol
Cunningham G, Katznelson L, 33:190201
Weber TJ, Friedman TC, Snyder P,
Metab 85:45004510 Zhang GY, Gu YQ, Wang XH, Cui YG,
Levine HL (2004) New testosterone
van der Vies J (1965) On the Bremner WJ (1998)
buccal system (Striant) delivers
mechanism of action of nandrolone A pharmacokinetic study of
physiological testosterone levels:
phenylpropionate and nandrolone injectable testosterone undecanoate
pharmacokinetics study in
decanoate in rats. Acta Endocrinol in hypogonadal men. J Androl
hypogonadal men. J Clin Endocrinol
49:271282 19:761768
Metab 89:38213829
van der Vies J (1970) Model studies in Zitzmann M (2009) Pharmacogenetics
Wang C, Ilani N, Arver S, McLachlan
vitro with long-acting hormonal of testosterone replacement
RI, Soulis T, Watkinson A (2011)
preparations. Acta Endocrinol therapy. Pharmacogenomics
Efficacy and safety of the 2%
64:656669 10:13411349
formulation of testosterone topical
van der Vies J (1985) Implications of basic solution applied to the axillae in Zitzmann M, Nieschlag E (2007)
pharmacology in the therapy with androgen-deficient men. Clin Androgen receptor gene CAG
esters of nandrolone. Acta Endocrinol Endocrinol (Oxf) 75:836843 repeat length and body mass index
Suppl (Copenh) 271:3844 modulate the safety of long-term
Wang L, Shi DC, Lu SY, Fang RY (1991)
intramuscular testosterone
Vest SA, Howard JE (1939) Clinical The therapeutic effect of
undecanoate therapy in
experiments with androgens. IV: a domestically produced testosterone
hypogonadal men. J Clin Endocrinol
method of implantation of undecanoate in Klinefelter
Metab 92:38443853
crystalline testosterone. J Am Med syndrome. New Drugs Mark 8:2832
Assoc 113:18691872 Zitzmann M, Hanisch JU, Mattern A,
Werner SC, Hamger FM, Kritzler RA
Maggi M (2011) Testosterone
von Eckardstein S, Nieschlag E (2002) (1950) Jaundice during
replacement therapy in male
Treatment of male hypogonadism methyltestosterone therapy. Am
hypogonadism: Final results from
with testosterone undecanoate J Med 8:325331
the largest international
injected at extended intervals of 12 Westaby D, Ogle SJ, Paradinas FJ, substitution trial involving 1493
weeks: a phase II study. J Androl Randell JB, Murray-Lyon IM (1977) patients [abstract]. Endocr Rev
23:419425 Liver damage from long-term 32 (03_MeetingAbstracts):P1347

335
Chapter
Androgens in male senescence

16 Jean-Marc Kaufman, Guy TSjoen, and Alex Vermeulen

16.1 Introduction 336 16.4 Clinical relevance of hypoandrogenism of


16.2 Declining endocrine testicular function in senescence 345
senescence 337 16.4.1 General background 345
16.2.1 Testosterone production and serum 16.4.2 Hypoandrogenism of senescence and
levels 337 sexual activity 346
16.2.2 Sex hormone-binding globulin and 16.4.3 Testosterone and cardiovascular risk
free testosterone serum levels 338 profile 347
16.2.3 Tissue levels and metabolism of 16.4.4 Body composition and sarcopenia 348
androgens 339 16.4.5 Senile osteoporosis 349
16.2.4 Factors affecting serum testosterone 16.4.6 Additional clinical variables 351
levels in elderly men 340 16.4.7 Conclusions 352
16.2.4.1 Influence of physiological factors 16.5 Androgen therapy in elderly men 352
and lifestyle 340
16.5.1 Who should be considered for
16.2.4.2 Testosterone serum levels in
treatment? 352
disease 342
16.5.2 Potential benefits 355
16.3 Physiopathology of declining testosterone
16.5.3 Potential risks 355
levels in senescence 343
16.5.4 Modalities of androgen
16.3.1 Primary testicular changes 343
substitution 357
16.3.2 Altered neuroendocrine regulation 344
16.6 Key messages 357
16.3.3 Increase of serum sex hormone-binding
16.7 References 358
globulin 345

16.1 Introduction population means for semen parameters are limited


Andropause, defined as the male equivalent of the to a decrease of ejaculate volume and sperm motility,
menopause, which in women signals the end of reproduct- with a decrease in total sperm output and percentage
ive life and a near total cessation of sex steroid production spermatozoa with normal morphology reported in
by the gonads, does not exist. Indeed, aging in healthy men some studies (Rolf et al. 1996; Ng et al. 2004; Lazarou
is normally not accompanied by abrupt or drastic alter- and Morgentaler 2008). Moreover, decreased ejacula-
ations of gonadal function, and androgen production as tory frequency, as observed in elderly men (Rolf et al.
well as fertility can be largely preserved until very old age. 1996), might account for at least part of these age-
The limited data available suggests that aging has related changes (Cooper et al. 1993). Serum inhibin B,
limited influence on sperm quality and fertilizing a marker of Sertoli cell function and spermatogenesis,
capacity (Nieschlag et al. 1982; Rolf et al. 1996; Jung was shown to be relatively well maintained in healthy
et al. 2002). Most consistently reported changes of elderly men, albeit at the cost of clearly increased FSH
stimulation that compensates for an age-related

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

336
Chapter 16: Androgens in male senescence

regression of Sertoli cell mass and function nmol/l


(Mahmoud et al. 2000). 70
Concerning hormonal testicular function, it is now Testosterone
60 FT (100)
well established that mean serum testosterone levels
SHBG
decrease progressively in generally healthy elderly 50
men, even though there is a considerable interindivi-
dual variability in the extent of the changes (Kaufman 40
and Vermeulen 2005). Well over 20% of otherwise 30
apparently healthy men over 60 years of age present
with decreased testosterone levels compared to serum 20
levels in young adults. Moreover, this age-dependent
10
decline in androgen production can be accentuated by
co-morbidity, with transient or more permanent 0
adverse effects on Leydig cell function (Kaufman and 2534 3544 4554 5564 6574 7584 85100
Vermeulen 2005; Travison et al. 2007). The relative Age (years)
contribution of aging versus subtle co-morbidities as Fig. 16.1 Mean serum levels of testosterone, free testosterone (FT)
cause of the declining mean serum testosterone levels and sex hormone-binding globulin (SHBG) according to age in a
in study populations of apparently healthy aging men cross-sectional study of 300 healthy men (according to data in
Vermeulen et al. 1996).
inevitably remains an area of some controversy.
The extent to which a relative hypoandrogenism in
the elderly contributes to clinical signs and symptoms Whether aging in healthy men is also associated
of aging remains an issue not fully explored but deserv- with decreased serum testosterone concentrations has
ing further attention, as many clinical features of aging long been a controversial issue. But a decline of serum
in men are reminiscent of those of hypogonadism in levels from the fourth or fifth decade of life on, or
younger subjects. Accumulating evidence indicates that even earlier according to some reports, has been
clearly decreased serum testosterone levels in elderly demonstrated in a series of cross-sectional studies
men predict poorer health and survival. The indications which included healthy ambulatory young and elderly
for androgen supplementation to aging men, as well as men sampled in the morning (Vermeulen 1991; Kauf-
its potential merits, remain a subject of debate. man and Vermeulen 2005; Mohr et al. 2005 for
review). The observed mean serum testosterone
16.2 Declining endocrine testicular values at age 75 years are about two-thirds of those
at age 25 years (Fig. 16.1).
function in senescence An age-related decrease of serum testosterone
16.2.1 Testosterone production and levels has also been documented in longitudinal stud-
ies (Morley et al. 1997; Zmuda et al. 1997; Harman
serum levels et al. 2001; Feldman et al. 2002; Andersson et al. 2007;
Early reports of decreased spermatic vein testosterone Travison et al. 2007; Liu et al. 2007a; Lapauw et al.
blood concentrations (Hollander and Hollander 1958) 2008). In fact, the longitudinally assessed decline of
and decreased testosterone blood production rates serum testosterone tends to be even larger than appar-
(Kent and Acone 1966) in elderly men compared to ent from cross-sectional analyses, which might be due
younger individuals were subsequently confirmed by to a bias towards healthier subjects in the latter, while
several studies performed in the seventies (Vermeulen during longitudinal follow-up more elderly subjects
et al. 1972; Giusti et al. 1975; Baker et al. 1977). are likely to show deterioration than an improvement
However, the observation of reduced testosterone of their general health status relative to the baseline
blood production rate does not necessarily imply situation.
lower testosterone plasma levels. Indeed, the blood Whereas mean serum testosterone levels in the
production rate is the product of the mean plasma adult male population decrease with age, at all ages
levels and the metabolic clearance rate, whereby the there is a large interindividual variability of serum
latter is also reduced in elderly men (Kent and Acone testosterone levels, with some elderly men having
1966; Vermeulen et al. 1972; Coviello et al. 2006). frankly low serum testosterone levels while many

337
Chapter 16: Androgens in male senescence

n = 40
et al. 2009). These findings illustrate that in
40 unselected populations the age-related decrease of
Total T < 11 nmol/l
total serum testosterone seen in apparently healthy
Free T < 0.18 nmol/l men tends to be masked by a higher prevalence of
30 low testosterone in younger age groups.
n = 87
Percentage

20 16.2.2 Sex hormone-binding globulin


and free testosterone serum levels
Whereas some authors may still argue that total tes-
10 n = 68
tosterone concentrations are not reduced in perfectly
healthy elderly men, there is virtually unanimity
n = 105
0
amongst authors that the free and non-specifically
2040 4060 6080 >80
bound serum testosterone, which is generally con-
sidered to represent the serum testosterone fractions
Age (years) readily available for biological activity, does indeed
Fig. 16.2 Proportion of healthy men presenting with decrease with age (Kaufman and Vermeulen 2005 for
subnormal serum levels for total testosterone (Total T <11 nmol/l) or
free testosterone (Free T <0.18 nmol/l) (from Kaufman and
review). In healthy ambulatory men, mean serum
Vermeulen 1997). levels of free testosterone and of non-SHBG-bound
or so-called bioavailable testosterone (i.e. the sum
of the free fraction and the fraction loosely bound to
others have perfectly preserved testosterone secretion, albumin) decrease by as much as 50% between the
with serum levels well within the normal range for ages of 25 and 75 years (Vermeulen et al. 1996; Ferrini
young adults. With advancing age, a progressively and Barrett-Connor 1998). The sharper decline of
larger proportion of men present with subnormal these fractions in comparison with total testosterone
values relative to those in young adults; in a group is explained by an age-associated increase of SHBG
of 300 healthy men aged 20 to 100 years (Vermeulen concentrations, and confirmed in longitudinal studies
et al. 1996), we observed a subnormal testosterone (Morley et al. 1997; Harman et al. 2001; Feldman
level in less than 1% of men below age 40 years et al. 2002; Andersson et al. 2007; Travison et al.
but in more than 20% of men older than 60 years 2007; Lapauw et al. 2008). In 300 healthy men aged
(Fig. 16.2). In an unselected, population-based sample 25100 years we observed an approximately log linear
of the Third National Health and Nutrition Examin- decrease of free testosterone levels at a rate of 1.2%
ation Survey (NHANES III; USA), the prevalence of per year (Fig. 16.1); while total serum testosterone
men with low serum testosterone defined as below remained relatively stable up to the age of 55 years
300 ng/dl (10.4 nmol/l) was 25% (Rohrmann et al. and declined thereafter at a rate of 0.85% per year
2011). Also in an unselected, population-based (Vermeulen et al. 1996). The decline in serum testos-
sample, Araujo et al. (2007) observed a prevalence of terone persists in old age. In a group of apparently
over 20% of low total testosterone (<300 ng/dl) in healthy older men (n = 218) with population-based
men over 60 years of age, but in this study the preva- recruitment, aged 71 to 86 years at baseline, the per-
lence was rather similar in younger men aged 40 to 60 centual yearly decline assessed longitudinally over
years, which can be explained by the presence of co- four years amounted to 1.26%, 1.33% and 2.43% for
morbidity and mainly a high prevalence (>30%) of total, free and non-SHBG-bound testosterone,
obesity in this study population. A similar proportion respectively (Lapauw et al. 2008).
of men over 65 years with serum testosterone below As is the case for total testosterone, there is a great
300 ng/dl, without further age-related changes in this between-subject variability in prevailing free (or bio-
age-group, was reported for the large unselected available) testosterone levels in elderly men, ranging
population in the MrOS study (Orwoll et al. 2006a). from markedly low levels to levels in the upper normal
In an unselected German population of primary care range for young adults (Fig. 16.3), the proportion of
patients with mean age of 59 years, 20% presented men with subnormal free testosterone levels increasing
with serum testosterone below 300 ng/dl (Schneider with age (Fig. 16.2). As a result of the more rapid

338
Chapter 16: Androgens in male senescence

(A) Fig. 16.3 Histogram for the distribution


of serum free testosterone in (A) 353
80
community-dwelling elderly men without
major health problems, aged 70 to 85
years and (B) in a younger control
Number of subjects

60 population of healthy men aged 23 to 58


years; the lower limit for the laboratory
normal range is indicated by the arrows.
40

20

0
(B)
20
Number of subjects

15

10

0
0 4 8 12 16 20 24 28
Serum free testosterone (ng/ml)

decline of free testosterone, the prevalence of elderly Androgen receptors are present at highest concen-
men with low free testosterone is greater than for low trations in the accessory sex organs; whereas concen-
total testosterone (Kaufman and Vermeulen 2005; trations in other androgen-sensitive tissues such as
Rohrmann et al. 2011). However, as discussed below, the heart and bone are much lower. Tissue concen-
limits of normality are somewhat arbitrary as the sen- trations both of androgen receptors (Rajfer et al.
sitivity threshold for androgen action may vary from 1989) and of androgens decrease with age (Deslypere
tissue to tissue and according to age. and Vermeulen 1981; 1985).
Overall, the influence of the aging process on the
16.2.3 Tissue levels and metabolism metabolism of testosterone manifests itself essentially by
a decrease of the metabolic clearance rate (Vermeulen
of androgens et al. 1972; Coviello et al. 2006), which results from an
It is generally considered that free testosterone diffuses age-associated decrease of cardiac output and of hepatic
passively within the cell. It should be mentioned, how- as well as tissue perfusion, and from increased binding of
ever, that during transit through the capillaries, a large plasma testosterone to SHBG.
part of the albumin-bound testosterone dissociates and Of major physiological significance is the activat-
becomes bioavailable. Testosterone bound by SHBG is ing biotransformation in the tissues, with testoster-
not directly available to the cells, but the physiopathol- one acting as a prohormone and being metabolized
ogy of SHBG is still incompletely understood. It has into the more potent androgen DHT by type 2 or
been shown that in some tissues (e.g. in the prostate type 1 5a-reductase, and to estrogens by the
and epididymis) cellular membranes carry SHBG cytochrome P450 aromatase enzyme. Production of
receptors (Rosner et al. 1991; Porto et al. 1992), the DHT occurs in, or in the close vicinity of the target
activation of which stimulates intracellular cAMP pro- cells, as does also its catabolic metabolism, with only
duction. Moreover, the SHBGandrogen complex a minor fraction escaping to the general circulation;
might be internalized by several cell types. whereas DHT produced in the liver is locally

339
Chapter 16: Androgens in male senescence

glucuronoconjugated and does not reach the general


circulation. Blood production rate thus largely 16.2.4 Factors affecting serum
underestimates total DHT production in the organ-
ism. Aromatization of testosterone in (subcutaneous)
testosterone levels in elderly men
fat and in striated muscle is the major contribution 16.2.4.1 Influence of physiological factors
to circulating estradiol in men; both estradiol in the and lifestyle
systemic circulation and estradiol locally produced in The physiological basis underlying the large interin-
the target tissues can contribute to estrogen-medi- dividual variation in serum testosterone levels seen at
ated testosterone effects. The expression both of 5a- any age is not yet fully elucidated, but several physio-
reductase activity and of the aromatase enzyme logical variables and factors related to lifestyle have
coded by the CYP19 gene can be differentially regu- been identified that account for part of the wide range
lated in different target tissues (Russell and Wilson of normal values observed in healthy men. In any
1994; Kamat et al. 2002), local biotransformation to case, the apparent interindividual variability of testos-
DHT and estrogens being potentially important terone levels is not merely artefactual as a result of a
regulatory components in the modulation of tissue cross-sectional design of the clinical studies, as single-
effects of testosterone. point plasma testosterone estimates reflect, fairly
No data are available concerning the possible well, longer-term androgen status in healthy men
influence of aging on blood conversion rates of tes- (Vermeulen and Verdonck 1992).
tosterone to DHT. Reports on age-related changes of The circadian variation of serum testosterone,
DHT serum levels are not consistent, but do not with highest levels in the early morning and lowest
suggest major changes, and in any case serum DHT levels in the late afternoon, should not play an
is not a reliable parameter of tissue DHT formation important role in the wide range of normal testoster-
(Labrie et al. 1995). one levels if they are consequently evaluated in the
The conversion of testosterone to estradiol morning (preferably before 10 a.m.), as should be the
increases with age (Siiteri and MacDonald 1975; rule. However, recent reports suggest that serum tes-
Lakshman et al. 2010), which appears to be the tosterone is higher in fasting state (Sartorius et al.
consequence of an increase of both aromatase 2011). The ultradian pattern of episodic testosterone
tissue activity and of fat mass in the elderly (Ver- secretion undoubtedly contributes to the variability
meulen et al. 1996; Vermeulen and Kaufman 2002). of testosterone levels (Veldhuis et al. 1987; Spratt et al.
This increasing global aromatase activity compen- 1988). Therefore, although single time-point esti-
sates for the decreased substrate availability, i.e. the mates are a valid approach for clinical studies, the
declining testosterone and androstenedione plasma existence of fluctuations in serum testosterone
levels, so that the serum estradiol levels do not should be taken into account when assessing the
show much change or only slight decline during androgen status of individual elderly subjects (Morley
aging in men (Vermeulen et al. 1996; Ferrini and et al. 2002). The occurrence of circannual variations
Barrett-Connor 1998; Vermeulen and Kaufman with amplitudes of up to 35% have been reported,
2002; Orwoll et al. 2006a). However, there is a with the highest testosterone levels measured between
decrease with age of the serum testosterone-to- October and December for studies performed in the
estradiol ratio and, as a consequence of the age- western hemisphere (Smals et al. 1976; Svartberg et al.
related increase in SHBG, also a moderate decrease 2003). However, other studies failed to find such
of serum free and bioavailable estradiol (FE2 variations or observed a maximum rather in spring
and Bio E2). or summer (Svartberg et al. 2003; Ruhayel et al. 2007;
Serum levels of 5a-androstane-3a,17b-diol- Brambilla et al. 2007).
glucuronide (ADG) decrease significantly with age Heredity appears to play an important role, as
(Deslypere et al. 1982), a consequence of the concluded from studies in twins and siblings (Meikle
decreased serum concentrations of the precursors et al. 1986; 1988; Ring et al. 2005; Bogaert et al. 2008),
(70% testosterone and 30% DHEAS). A decrease of with up to 60% of the variability of serum testosterone
the ratio of 5a/5b metabolites in urine indicates an attributable to genetic factors. Nevertheless, non-
age-related decrease of type 2 5a-reductase activity genetic factors also have a substantial impact on
(Vermeulen et al. 1972; Zumoff et al. 1976). testosterone serum levels.

340
Chapter 16: Androgens in male senescence

The genetic basis for the heredity of serum (free) In clinical studies, body composition and, in par-
testosterone and SHBG is presently largely unknown. ticular, adiposity emerges as an important determin-
Reports of ethnic differences in serum testosterone ant of SHBG levels. For the whole range of clinically
levels have been inconsistent, with small differences encountered BMI values there is a highly significant
tending to disappear if adjustments are made for negative correlation with SHBG and testosterone
differences in body composition, especially for serum levels, which is explained at least in part by
abdominal adiposity (Winters et al. 2001; Gapstur increased insulin levels (Khaw and Barrett-Connor
et al. 2002; Heald et al. 2003; Rohrmann et al. 1992; Giagulli et al. 1994; Yeap et al. 2009a). In elderly
2007). Recently, regional and racial differences in men this inverse relationship between BMI and SHBG
sex steroids were reported for elderly men in the levels as well as total testosterone can be clearly dem-
large MrOS study. Serum testosterone was 20% onstrated notwithstanding the background of an age-
higher in Asian men living in Hong Kong or Japan, related rise of SHBG levels (Vermeulen et al. 1996;
but not in the USA compared to the other groups. Mohr et al. 2006; Orwoll et al. 2006a; Wu et al. 2008).
Again these differences tended to disappear, at least Similarly, a negative association of serum SHBG and
for total testosterone, when adjusted for BMI total testosterone with leptin levels has been observed
(Orwoll et al. 2010). in elderly men (Haffner et al. 1997; Van Den Saffele
There has been considerable interest for the role of et al. 1999). Negative associations with serum testos-
a polymorphic trinucleotide CAG-repeat contained in terone levels tend to be most pronounced for indices
exon 1 of the AR, which encodes a functionally rele- of abdominal adiposity (Khaw and Barrett-Connor
vant polyglutamine tract of variable length. A CAG- 1992; Vermeulen et al. 1999a; Couillard et al. 2000;
repeat length exceeding the normal range of 1531 Kupelian et al. 2008; Hall et al. 2008; Grossmann
results in diminished AR transactivation capacity. 2011). Whereas in younger men moderate obesity
There is evidence that androgen activity is also mainly affects total serum testosterone by lowering
affected by variations in AR gene CAG-repeat length SHBG binding capacity, with decreased free testoster-
within the normal range: shorter repeats being asso- one seen in morbid obesity (BMI > 3540) as a result
ciated with increased AR transactivation capacity and of neuroendocrine disturbances (Giagulli et al. 1994),
clinical correlates of increased androgen activity elderly men might be even more sensitive to the
(Zitzmann and Nieschlag 2003). Several studies have negative effects of adiposity on free or bioavailable
shown positive associations of serum total and free- testosterone (Vermeulen et al. 1999a; Van Den Saffele
testosterone levels with AR CAG-repeat length, which et al. 1999; Couillard et al. 2000; Mohr et al. 2006). In
likely reflect the effect of the polymorphism on andro- this context it is interesting to note that Andersson
gen sensitivity and consequently on the negative feed- et al. (2007) described a birth cohort effect with
back set point for serum testosterone (Crabbe et al. higher serum total testosterone, but not free testoster-
2007; Stanworth et al. 2008; Huhtaniemi et al. 2009; one, in older generations resulting from a decline of
Lindstrm et al. 2010). The effect of this AR poly- SHBG in more recent generations. This appears to be
morphism on serum testosterone may be absent or explained by a parallel increase of BMI in the latter
reduced in the more elderly; possibly because of generations. When discussing the effects of adiposity
altered negative feedback regulation (Van Pottelbergh on serum androgens one should keep in mind the
et al. 2003a; Harkonen et al. 2003). Other polymorph- caveat that, as discussed later, the relation is bidirec-
isms in the androgen receptor (Bogaert et al. 2009) tional, with low serum testosterone favoring increased
and other genes involved in the pituitary-gonadal adiposity.
function (Eriksson et al. 2006; Vanbillemont et al. Overt alterations in thyroid hormone levels have
2009; Huhtaniemi et al. 2010) have been reported to marked effects on SHBG levels, with thyrotoxicosis
affect testosterone levels. resulting in a several-fold increase of SHBG levels and
Several metabolic and hormonal factors influence marked increase of total serum testosterone (Vermeu-
SHBG serum levels. Insulin and insulin-like factor len et al. 1971). More subtle changes in thyroid hor-
I (IGF-1) inhibit SHBG production and, in clinical mone levels can also affect SHBG and testosterone
studies, insulin was found to be inversely correlated levels, with significant increases of SHBG and testos-
with serum SHBG and testosterone levels (Haffner terone levels seen in subclinical hyperthyroidism,
et al. 1988; Vermeulen et al. 1996). with suppressed serum thyroid-stimulating hormone

341
Chapter 16: Androgens in male senescence

without clinical hyperthyroidism or elevation of thy- this will remain controversial as it may not be realistic
roid hormone levels above normal (Faber et al. 1990; to definitely exclude subtle co-morbidities in appar-
Giagulli and Vermeulen 1992). ently healthy older men without selecting out a poorly
Concerning lifestyle factors, reports on the effects of representative group of super-healthy elderly. In
diet on serum testosterone levels have not always been any case, diseases and/or their treatment can be asso-
concordant, but it seems possible to conclude that diet ciated with transient or more permanent decreases of
influences testosterone levels mainly indirectly through testosterone production (Handelsman 1994; Turner
changes in SHBG levels, fiber-rich, vegetarian diets and Wass 1997). Considering the high prevalence of
being associated with higher SHBG and testosterone diseases and use of medication that can adversely
levels than western-type diets with high fat content. affect testosterone levels in the elderly, it is evident
Further, low protein intake is associated with decreased that in daily practice the age-associated decline in
IGF-1, which in turn may result in increased SHBG and testosterone levels may often be accentuated by inter-
total testosterone (Kaufman and Vermeulen 2005 for current disease (Mohr et al. 2005; Orwoll et al. 2006b;
review). In men aged 40 to 70 years in the Massachusetts Travison et al. 2007; Schneider et al. 2009).
Male Aging Study, fiber intake and protein intake, but Acute critical illness, such as acute myocardial
not carbohydrate, fat or total caloric intake, were inde- infarction or surgical trauma, is associated with
pendent positive and negative determinants, respect- temporary, but often profound and prolonged
ively, of serum SHBG (Longcope et al. 2000). decrease of serum (free) testosterone (Wang et al.
Current smoking at all ages has generally been 1978; Woolf et al. 1985; Swartz and Young 1987;
associated with 5 to 15% higher total and free serum Spratt et al. 1993; Impallomeni et al. 1994; Vanden-
testosterone levels than in non-smokers (Barrett- berghe et al. 2001).
Connor and Khaw 1987; Dai et al. 1988; Field et al. A series of chronic diseases can induce more long-
1994; Vermeulen et al. 1996). In the European Male standing decreases in testosterone levels (Handelsman
Ageing Study (EMAS), including men 40 to 79 years 1994; Turner and Wass 1997). Both testosterone and
without exclusion of co-morbidities, smoking was SHBG levels tend to be decreased in elderly men with
associated with increased serum levels of LH, SHBG insulin resistance, metabolic syndrome and type 2
and total, but not free, testosterone (Wu et al. 2008). diabetes (Andersson et al. 1994; Kupelian et al. 2008;
Alcohol abuse, also in the absence of liver cirrhosis, Yeap et al. 2009a; Grossmann 2011).
may accentuate the age-associated decrease of testos- Coronary atherosclerosis has been reported to be
terone levels, estradiol serum levels being increased accompanied by lower or similar testosterone levels as
(Cicero 1982; Irwin et al. 1988; Ida et al. 1992); compared to controls (Poggi et al. 1976; Lichtenstein
moderate alcohol consumption has no adverse effect et al. 1987; Swartz and Young 1987; Phillips et al.
(Sparrow et al. 1980; Longcope et al. 2000). 1994; Alexandersen et al. 1996; Hak et al. 2002).
Both physical and psychological stress and strenu- Low serum testosterone has also been associated with
ous physical activity result in transient or more pro- carotid artery atherosclerosis, incident stroke and
longed decreases of serum (free) testosterone aneurisms of the aorta abdominalis (van den Beld
(Kaufman and Vermeulen 2005 for review); although et al. 2003; Yeap et al. 2009b; 2010). However, it is
elderly men have been reported to be more resistant not clear whether decreased testosterone levels repre-
to stress such as the metabolic stress of fasting (Ber- sented a consequence of atherosclerosis or rather a
gendahl et al. 1998). pre-existent risk factor for cardiovascular disease (see
Low serum (free) testosterone has also been asso- Wu and von Eckardstein 2003 for review).
ciated with low income and with recent loss of spouse In COPD and in patients with other hypoxic pul-
(Travison et al. 2007; Hall et al. 2008). Whether sexual monary diseases, serum testosterone levels are often
activity influences serum testosterone levels remains a decreased with inappropriately low gonadotropin
controversial issue as limited literature data are not levels, also in the absence of systemic glucocorticoid
consistent. treatment (Semple et al. 1984; Kamischke et al. 1998).
Sleep apnea syndrome is commonly accompanied by
16.2.4.2 Testosterone serum levels in disease relative hypogonadotropic hypogonadism, potentially
There is considerable evidence that the aging process reversible by nasal continuous positive airways pres-
per se adversely affects testosterone production, but sure therapy; massive obesity is often a contributing

342
Chapter 16: Androgens in male senescence

factor (Worstman et al. 1987; Grunstein et al. 1989; unchanged or modestly elevated (Vermeulen et al.
Luboshitzky et al. 2002). 1989a), but treatment can result in mild symptoms
Chronic renal failure is often accompanied by of hypogonadism (Thompson et al. 2003) by mitigat-
hypogonadism with complex pathophysiology, involv- ing androgen effects in those tissues where andro-
ing the different compartments of the hypothalamo- genic effects are largely mediated by DHT.
pituitary-gonadal axis (Handelsman and Dong Spironolactone can reduce testosterone production
1993; Veldhuis et al. 1993). In chronic liver disease, through an inhibitory action on the 17-hydroxylase-
decreased (free) testosterone levels are accompanied lyase activity (Stripp et al. 1975).
by increased SHBG, androstenedione and estrone Several classes of antihypertensive drugs, includ-
levels (Baker et al. 1979; Elewaut et al. 1979). Hypogo- ing b-blockers, may adversely affect erectile function,
nadism in hemochromatosis is multifactorially and hypertensive patients under treatment may show
determined, with a major contribution of pituitary mildly decreased serum testosterone levels (Turner
insufficiency (Duranteau et al. 1993). and Wass 1997); considering the many confounding
Moderate impairment of testicular function has factors in these patients, an adverse effect of antihy-
been observed in active rheumatic diseases (Tapia- pertensive treatment on serum testosterone is not to
Serrano et al. 1991). In the elderly, as in the young, be considered as established.
Leydig cell function may be adversely affected by
endocrine diseases such as Cushing syndrome and
pituitary tumors; in particular, prolactinomas. There 16.3 Physiopathology of declining
is substantial overlap between symptoms of hypogo- testosterone levels in senescence
nadism and hypothyroidism, which itself is a possible
cause of hypogonadism (Brenta et al. 1999). 16.3.1 Primary testicular changes
Finally, in the elderly there is a rather high preva- Primary testicular factors undoubtedly play an import-
lence of the use of medications, and concomitant use ant role in the age-associated decline of Leydig cell
of several drugs is far from exceptional. This may function, as indicated by a reduced absolute secretory
further negatively affect gonadal function already response to stimulation with hCG (Longcope 1973;
impaired by underlying disease. Such a situation is Nieschlag et al. 1973; 1982; Rubens et al. 1974; Harman
typically encountered in COPD patients treated with and Tsitouras 1980; Nankin et al. 1981). Diminished
glucocorticoids (Kamischke et al. 1998). Their testicular secretory capacity in the elderly has also been
chronic use may impair Leydig cell function, which confirmed for the response to recombinant human LH
induces a dose-dependent suppression of testosterone following downregulation of endogenous LH with
levels by combined actions at the testicular and at the leuprorelin (Mulligan et al. 2001), and for the response
hypothalamo-pituitary level, and by a decrease of to prolonged physiologic intermittent stimulation
SHBG serum levels (MacAdams et al. 1986; by endogenous LH during a two-week pulsatile GnRH
Kamischke et al. 1998). Opiates can induce hypogo- infusion (Mulligan et al. 1999). This decrease in tes-
nadotropic hypogonadism (Daniell 2002; Rajagopal ticular reserve for testosterone secretion appears to
et al. 2004). Secretion of LH and Leydig cell function result from a reduced number of Leydig cells (Sniffen
may be adversely affected by hyperprolactinemia 1950; Harbitz 1973; Neaves et al. 1984). There is,
during chronic use of neuroleptic drugs and related moreover, evidence for involvement of vascular
compounds (Bixler et al. 1977). Leydig cell function changes (Sasano and Ichijo 1969; Suoranta 1971), the
can be mildly affected by cytotoxic chemotherapy deficient oxygen supply inducing changes in testicular
(Howell et al. 2001). steroid metabolism (Pirke et al. 1980; Vermeulen and
If only as a reminder, one has to mention hormo- Deslypere 1986). In healthy community-dwelling eld-
nal treatment of prostate cancer, which is aimed at erly men 70 years or older, mean testicular volume is
inducing profound hypogonadism by suppression of decreased by about 30% as compared to young men
LH and testosterone secretion by use of a GnRH (Mahmoud et al. 2003).
analog and/or by blockade of androgen effects with In apparent agreement with the view of a primary
an antiandrogen. As to the use of 5a-reductase testicular defect are observations of moderate
inhibitors in benign prostate hypertrophy, under increases of basal gonadotropin levels in elderly men
treatment with finasteride testosterone levels are as observed in several, but not all, studies on the age-

343
Chapter 16: Androgens in male senescence

related decline of testicular function (Vermeulen near physiologic doses of synthetic GnRH has
1991; Tsitouras and Bulat 1995 for review). Although revealed a preserved or, in accordance with a status
there have been some discordant findings (Urban of relative hypoandrogenism, even moderately
et al. 1988; Mitchell et al. 1995), this increase is not increased LH response in the elderly compared to
limited to immunoreactive forms of the gonadotro- the young (Kaufman et al. 1991; Mulligan et al.
pins, as it is also demonstrated for levels measured by 1999). If the pituitary secretory capacity is main-
bioassay (Tenover et al. 1988; Kaufman et al. 1991; tained, hypothalamic mechanisms must be involved
Matzkin et al. 1991). Part of the age-related increase in the apparent failure of the feedback regulation to
of gonadotropin levels may result from reduced produce an adequate LH rise in response to hypoan-
plasma clearance (Kaufman et al. 1991; Bergendahl drogenism (Vermeulen and Kaufman 1992).
et al. 1998). The increase of LH with age is usually of Several changes in the neuroendocrine control of
rather modest amplitude and inconsistent (Morley Leydig cell function likely situated at the hypothal-
et al. 1997), but may be more prominent when con- amic level have been documented in elderly men.
sidering populations with substantial prevalence of Circadian rhythm of LH and testosterone secretion
co-morbidity (Wu et al. 2008; Tajar et al. 2010). In a is clearly blunted in elderly men (Bremner et al. 1983;
longitudinal assessment in community-dwelling men Tenover et al. 1988; Plymate et al. 1989). Further-
over age 70 years, both serum LH and FSH were more, the pulsatile pattern of LH secretion controlled
independent predictors of serum (free) testosterone by intermittent secretion of GnRH is altered, with
decrease (Lapauw et al. 2008). increased irregularity of LH pulses (Pincus et al.
1997; Veldhuis et al. 2000). Whereas mean LH pulse
frequency remains essentially unchanged in the eld-
16.3.2 Altered neuroendocrine regulation erly (Winters et al. 1984; Tenover et al. 1987; 1988;
Although the combined observations of a diminished Urban et al. 1988; Vermeulen et al. 1989b), there is a
testicular reserve for testosterone secretion with diminished frequency of LH pulses with larger ampli-
increased basal gonadotropin levels reveal involve- tude and resulting decrease of the mean LH pulse
ment of a primary testicular factor in the age-related amplitude (Vermeulen et al. 1989b; Veldhuis et al.
decline of Leydig cell function, other mechanisms 1992), parameters of the stimulating effect on
must also be involved. Indeed, the responses to hCG the Leydig cells. Considering the preserved pituitary
challenges in elderly men indicate that the secretory LH secretory capacity, this decrease in LH pulse
reserve of the Leydig cells, albeit diminished, should amplitude must by inference be attributed to
still be sufficient to allow for a normalization of decreased stimulation by endogenous GnRH, with
plasma testosterone levels, provided the endogenous reduced size of the bolus of the neuropeptide inter-
drive by pituitary LH is adequate. The latter is indeed mittently released into the pituitary portal circulation.
the case in some of the elderly, who present with well- This could in turn result either from a decreased
maintained serum testosterone and elevated serum number of GnRH-secreting neurons, from their less
LH and are sometimes referred to as having compen- efficient intermittent recruitment and/or synchron-
sated hypogonadism (Tajar et al. 2010). However, ization, and/or from downregulation of their activity
many elderly men with serum testosterone below the by external local and/or systemic factors. This last
range for young men have normal or only slightly possibility is supported by evidence for increased
elevated serum LH. In the presence of a persistent responsiveness to the negative feedback effects of
status of relative hypoandrogenism, such LH levels testosterone in the elderly (Winters et al. 1984; 1997;
should be regarded as inappropriately low and indica- Deslypere et al. 1987; Mulligan et al. 1999). The latter
tive of alterations in the neuroendocrine regulation of might also underlie the fact that LH pulse frequency,
LH secretion. The relative inadequacy of the gonado- governed by the hypothalamic GnRH pulse generator
tropin response to hypoandrogenism in elderly men and expected to increase in a state of hypoandrogen-
has also been shown during experimental muting of ism, has been found by most authors to remain
endogenous testosterone suppression by administra- unchanged and thus inappropriately low; an increased
tion of an antiandrogen (Veldhuis et al. 2001). LH pulse frequency in elderly men having been
Assessment of the secretory capacity of the pituit- reported only by one group (Veldhuis et al. 1992;
ary gonadotropes by in-vivo challenge with small, Mulligan et al. 1995).

344
Chapter 16: Androgens in male senescence

The decreased hypothalamic drive of LH secretion 16.4 Clinical relevance of


in the elderly is not the consequence of an increased
inhibitory tone by endogenous opioids, which on the hypoandrogenism of senescence
contrary is rather reduced (Vermeulen et al. 1989b; 16.4.1 General background
Mikuma et al. 1994), does not result from increased
In contrast with the menopause, the decline of androgen
inhibition by endogenous estrogens (TSjoen et al.
levels in men is slowly progressive until the eighth
2005a), or from relative leptin deficiency (Van Den
decade, with a substantial proportion of men maintain-
Saffele et al. 1999). The mechanisms underlying the
ing normal (free) testosterone levels. Frankly decreased
apparent relative deficiency of GnRH secretion in eld-
testosterone levels in elderly men are thus by far not a
erly men remain to be fully elucidated. Moreover, the
generalized phenomenon and, as fertility is also usually
role in their presentation of additional risk factors, such
well preserved until old age, there is no such phenom-
as increased adiposity, apart from aging per se, remains
enon as an andropause or male climacterium. Subnor-
a matter of debate (Wu et al. 2008; Sartorius et al. 2011).
mal serum testosterone with accompanying signs and
symptoms of androgen deficiency in aging men is pres-
16.3.3 Increase of serum sex hormone- ently most commonly referred to as late-onset
binding globulin hypogonadism (LOH) (Wang et al. 2008). A key ques-
tion is the clinical relevance of the apparent partial
The progressive increase of plasma SHBG binding
androgen deficiency occurring in some elderly men.
capacity with age should be regarded as a third
Many of the clinical features of aging in men are
important aspect of the pathophysiological mechan-
reminiscent of the clinical changes seen in hypogo-
isms responsible for the age-related changes in circu-
nadism in young men, with a decrease in general well-
lating testosterone levels. Indeed, against the
being, mood changes, a decrease of energy and viril-
background of a relative inability to respond opti-
ity, a decrease in sexual pilosity and skin thickness,
mally to hypoandrogenism by increased testosterone
a decrease in muscular mass and strength, an increase
secretion in the elderly, due to the aforementioned
in upper and central body fat, a decrease in bone
testicular and neuroendocrine alterations, an inde-
density and increased prevalence of osteoporotic frac-
pendent progressive increase of SHBG binding cap-
tures, insomnia, sweating and hot flushes, a decrease
acity results in a steeper decline of the non-specifically
in libido and sexual drive, and markedly increased
bound testosterone fractions, i.e. free and bioavailable
prevalence of ED. However, in the elderly this symp-
serum testosterone levels.
tomatology develops slowly and progressively, with
The substantial increase of SHBG concentrations
symptoms that are variable, subtle and not specific.
in elderly men (Vermeulen et al. 1996; Orwoll et al.
Hence, the clinical picture at best suggests the possi-
2006a; Liu et al. 2007a; Wu et al. 2008) is remarkable
bility of a hypoandrogenic state in elderly men, and it
as it occurs in the face of increased fat mass and
is equally plausible that many observed clinical
insulin levels factors that are known to be strong
changes and the age-related decrease in serum testos-
negative determinants of SHBG levels. But the cause
terone levels are coincident and independent conse-
of these increased SHBG levels remains unclear. It is
quences of aging.
unlikely that the decreased testosterone levels per se
Much available information is still derived from
are responsible, as the increase in SHBG levels is
cross-sectional studies, which revealed mostly only
observed at an earlier age than the decrease of testos-
weak correlations between androgen status and clin-
terone levels, and the estradiol serum levels are rather
ical parameters. This is not surprising as clinical
similar in young and elderly men (Vermeulen et al.
changes are often multifactorially determined, a com-
1996). Serum SHBG and testosterone levels have been
plex clinical picture arising from concurrent effects of
reported to be inversely correlated to 24-hour GH and
the aging process per se, a relative hypoandrogenism
IGF-1 levels (Erfurth et al. 1996; Pfeilschifter et al.
and intercurrent diseases. In elderly men there is a
1996; Vermeulen et al. 1996), and it has been pro-
striking interindividual variability not only of the
posed that decreased activity of the somatotropic axis
prevailing testosterone levels but also of the pace of
may play a role in the age-associated increase of
clinical aging. Furthermore, there is increasing evi-
SHBG levels and ensuing decrease of free testosterone
dence that androgen requirements may vary not only
levels (Vermeulen et al. 1996).

345
Chapter 16: Androgens in male senescence

between individuals but within the same person, with dose-response effect on libido and erectile function,
different signs and symptoms of hypogonadism asso- but not on parameters of sexual activity, in the eld-
ciated with different testosterone levels (Bhasin et al. erly, at variance with the absence of such a dose-
2005; Zitzmann et al. 2006; Wu et al. 2008). In any response effect in young men (Bhasin et al. 2001;
event, decreased androgen levels can at best be Gray et al. 2005). Several authors reported differences
responsible for part of the clinical changes observed in parameters of sexual desire or activity according to
in aging men. endogenous serum testosterone levels in the elderly
More recently an increasing amount of data (Tsitouras et al. 1982; Davidson et al. 1983; Schiavi
became available from prospective studies assessing et al. 1990; Travison et al. 2006; Zitzmann et al. 2006;
the relationship of sex steroid levels with various Araujo et al. 2007; Wu et al. 2010), but there is a
intermediary and clinical health outcomes, including broad overlap of serum testosterone levels in sexually
frailty and mortality in aging men. The difficulty in less or more active elderly men in these studies.
interpretation of these studies often is deciding Moreover, other studies failed to find an association
whether the hypoandrogenism is a specific and direct between androgen levels and the perception of sexual
cause of the health problem or rather a more general functioning (Perry et al. 2001; TSjoen et al. 2004).
marker of poorer health. Although potency and nocturnal penile tumes-
The studies examining the effects of androgen cence (NPT) require adequate testosterone levels,
treatment in elderly men provide precious informa- and although several studies show that hormonal
tion, but should also be interpreted with caution, as alterations might play some role in 6 to 45% of cases
the demonstration of beneficial effects of a pharma- (Morley 1986), most frequently the cause of impo-
cological intervention does not necessarily imply that tence in elderly males is non-hormonal. There exists,
the corrected clinical sign or symptom was due to pre- nevertheless, a correlation between bioavailable tes-
existent androgen deficiency. tosterone levels and the frequency of NPT (Morley
1986). Erections in response to visual stimuli, on the
16.4.2 Hypoandrogenism of senescence other hand, are largely testosterone independent in
normal subjects (Kwan et al. 1983; Bancroft 1984);
and sexual activity suggesting that whereas testosterone is required to
Aging in men is accompanied by a decrease in libido sustain NPT, it is much less required to maintain
and sexual activity. Mean coital frequency was response to external stimuli. Still, testosterone levels
reported to be about four times a week at age 2025 have been reported to correlate inversely with the
years and decreasing to less than twice a month latency of erection in response to visual stimuli
between age 75 and 80 years (Masters 1986; Tsitouras (Rubin et al. 1979; Lange et al. 1980). Testosterone
and Bulat 1995). stimulates the activity of the nitric oxide synthase
Whereas normal libido requires adequate testos- enzyme, which may explain reported synergistic
terone levels, as shown by the effect of testosterone effects between androgens and inhibitors of phospho-
withdrawal (Basaria et al. 2002) and that of replace- diesterase type 5 (Aversa et al. 2003).
ment therapy in hypogonadal males (Hajjar et al. Notwithstanding the rather mitigated picture of
1997; Snyder et al. 2000; Kunelius et al. 2002; Steidle the role of testosterone in the regulation of erectile
et al. 2003; Isidori et al. 2005a), the correlation of function, the manifest major contribution of non-
libido with plasma testosterone levels is rather poor; hormonal factors in the pathogenesis of ED in the
the testosterone concentration required to sustain elderly, and failure to observe an association between
sexual activity and maintain libido appears to be serum testosterone and erectile function in several
rather low, probably at or below the lower end of studies (Korenman et al. 1990; Feldman et al. 1994;
the normal range in young men (Gooren 1987; Bhasin Rhoden et al. 2002), more recent studies in elderly
et al. 2001), and there is evidence indicating that men reported increased prevalence of ED with
healthy adults have substantially higher androgen decreased serum (free) testosterone (Zitzmann et al.
levels than required for normal sexual behavior (Udry 2006; Wu et al. 2010). A dose-response effect of
et al. 1985; Buena et al. 1993). testosterone on erectile function was demonstrated
Nevertheless, in an experimental setting with in elderly men (Gray et al. 2005), and, in a meta-
graded testosterone replacement, there was a clear analysis of testosterone replacement treatment,

346
Chapter 16: Androgens in male senescence

a positive effect on erectile function was shown; how- testosterone has been shown to be inversely associated
ever, the treatment effects are rather modest, essen- with serum triglycerides, total cholesterol, LDL chol-
tially restricted to men with initially low or low- esterol (LDL-C), fibrinogen and plasminogen activa-
normal serum testosterone, and seen mostly in studies tor type 1. In a multivariate analysis in an occupation-
of shorter duration (Isidori et al. 2005b). based cohort of 715 middle-aged men, free testoster-
The relative contribution of testosterone itself one was most significantly positively associated with
and of its metabolites DHT and estradiol in the HDL-C and apolipoprotein B; while free estradiol was
maintenance of normal sexual function has not a stronger positive predictor of apolipoprotein E (Van
been extensively studied. At present there is no Pottelbergh et al. 2003b). Obviously, body compos-
conclusive evidence for a major role of estradiol ition and insulin sensitivity are important confoun-
(Davidson et al. 1983), even though estradiol may ders in the context of associations of serum sex
contribute to some aspects of male sexual behavior steroid concentrations with cardiovascular risk
(Carani et al. 1997). As to DHT, it can maintain factors (Wu and von Eckardstein 2003), and although
sexual function (de Lignieres 1993; Kunelius et al. in a number of studies associations between serum
2002), but 5a-reduction of testosterone to DHT testosterone and HDL-C tend to persist after adjust-
is not an absolute requirement, as indicated by ments for these confounders, the statistical correc-
only limited occurrence of sexual dysfunction tions may account only in part for their impact.
during treatment with the 5a-reductase inhibitor The metabolic syndrome and type 2 diabetes mel-
finasteride (Thompson et al. 2003). litus represent significant risk factors for CVD.
A number of cross-sectional studies have described
16.4.3 Testosterone and cardiovascular negative associations of serum (free) testosterone with
prevalence of (components of) metabolic syndrome
risk profile and type 2 diabetes (Grossmann 2011 for review).
There is a gender gap for cardiovascular disease Moreover, prospective studies have shown that low
(CVD) and cardiovascular mortality, with men being serum testosterone predicts the development of meta-
at higher risk. Extensive reviews have failed to provide bolic syndrome and diabetes (Rodriguez et al. 2007;
compelling evidence that this gender difference can be Kupelian et al. 2008; Grossmann 2011 for review),
explained on the basis of distinctive patterns of also in non-obese men (Kupelian et al. 2006).
endogenous sex hormone exposure in adulthood A previous extensive literature review (Wu and
(Liu et al. 2003; Wu and von Eckardstein 2003). von Eckardstein 2003), which included a large
Suppression of plasma testosterone levels in men number of cross-sectional studies as well as a small
with a GnRH analog increases HDL-C, an effect pre- number of then-available nested, case-control studies
vented by co-administration of testosterone (Gold- and prospective cohort studies, failed to establish a
berg et al. 1985; Moorjani et al. 1987; Bagatell et al. relationship between serum testosterone levels and
1992), which seems in accordance with the generally CVD; similarly, observational studies also failed to
held view that testosterone is responsible for a more establish a relationship between serum DHEA or its
atherogenic plasma lipid profile in postpubertal males sulfate (DHEAS) with CVD. A recent meta-analysis of
as compared to women in their reproductive years. 19 prospective, population-based cohort and nested
Experimental graded testosterone exposure in elderly case-control studies showed a weak negative associ-
men, resulting in hypogonadal to supraphysiological ation of serum total testosterone with CVD: estimated
levels, dose-dependently decreased HDL-C (Bhasin summary relative risk (RR) was 0.89 for a change of 1
et al. 2005). However, within a window situated standard deviation in total testosterone level (95% CI
around physiological serum testosterone levels in 0.83 to 0.96). Age of study population and year of
men, most authors have consistently observed a posi- publication modified the relationship between serum
tive association between endogenous serum testoster- testosterone and CVD. The estimated summary RR
one and HDL-C concentrations in healthy men (see was 1.01 (0.95 to 1.08) for studies including middle-
Bagatell and Bremner 1995; Wu and von Eckardstein aged men, and 0.84 (0.76 to 0.92) for studies includ-
2003 for review), which is the case not only for total ing men over 70 years of age; the latter studies showed
but also for free testosterone (Haffner et al. 1993; Van a particularly pronounced association if published
Pottelbergh et al. 2003b). Furthermore, serum after 1 January 2007. From this meta-analysis it was

347
Chapter 16: Androgens in male senescence

concluded that, in elderly men, endogenous testoster- related adverse events during transdermal testoster-
one may weakly protect against CVD or that, alterna- one treatment in a rather small group of elderly men
tively, low testosterone may indicate poor general with limited mobility and high prevalence of chronic
health (Ruige et al. 2011). In another recent meta- disease has raised concerns concerning the cardiovas-
analysis of community-based studies, partially over- cular safety of serum testosterone in subjects at higher
lapping with the first one, the lower tertile of serum risk (Basaria et al. 2010).
testosterone compared to the upper tertile was associ-
ated with 35% and 25% increased risk of all-cause
mortality (n = 11 studies) and CVD mortality (7 stud- 16.4.4 Body composition and sarcopenia
ies), respectively. Age, baseline serum testosterone, Aging in men is associated with a decrease of lean body
length of follow-up and whether blood was sampled mass and an increase of fat mass, especially in the
in the morning were factors contributing to substan- upper body and central body regions (Forbes and
tial heterogeneity between studies (Araujo et al. 2011). Reina 1970; Tenover 1994; Vermeulen et al. 1999a).
In conclusion, high testosterone levels within Fat mass increases from around a mean of 20% of body
the physiological range do not cause an atherogenic weight in young men to 30% or more in the elderly;
lipid profile. On the contrary, moderately low serum whereas muscle mass may decrease by as much as 35 to
testosterone levels are accompanied by lower HDL-C 40% from age 20 to 80 years (Bross et al. 1999). In a
and higher LDL-C levels. Nevertheless, whereas study in community-dwelling healthy men (Vermeu-
higher endogenous testosterone may confer a limited len et al. 1999a), we found a mean fat mass of 22.3% of
protective effect against CVD in the elderly, low body weight in 61 middle-aged men with mean age of
serum testosterone might rather represent a negative 42 years, as compared to 29.4% in 271 men with mean
indicator of general health status. age of 76 years, BMI being similar in both groups and
The effects of exogenous testosterone on cardio- lean body mass 30% lower in the elderly.
vascular risk factors are mixed with possibly deleteri- Fat mass, and in particular abdominal fat mass, is
ous changes such as reductions in HDL-C and negatively associated with serum (free) testosterone
apolipoprotein A1, besides potentially favorable levels (Vermeulen et al. 1999a; van den Beld et al.
changes such as a decrease in serum fibrinogen, 2000). However, the direction of this association
a reduction in abdominal fat mass, and augmented remains unclear, as low testosterone may be a positive
insulin sensitivity (Emmelot-Vonk 2008). According determinant of adiposity; whereas, conversely, adi-
to a meta-analysis, testosterone treatment moderately posity appears to be a negative determinant of serum
reduces fat mass and total cholesterol, in testosterone. Moreover, altered activity of the soma-
particular in men with initially low serum testoster- totropic axis may also play an important role in age-
one (<10 nmol/l), has no effect on LDL-C, and related changes in body composition. In any case,
reduces HDL-C, mainly in men with initially higher a negative association of free testosterone with fat
(>10 nmol/l) serum testosterone (Isidori et al. 2005a). mass in elderly men persists after correction for
There has been considerable interest in possible serum IGF-1 levels, which are positively correlated
beneficial direct vascular effects of androgens on to serum (free) testosterone and negatively to fat mass
coronary arteries in men with CAD, but this non- (Vermeulen et al. 1999a).
genomic action of testosterone may require rather In a majority of controlled trials of several months
high blood concentrations. Although the long-term duration with administration of androgens to elderly
effects of testosterone treatment on risk for CAD in men with low or (low) normal serum (bioavailable)
elderly men are not established and the available testosterone, treatment resulted in a modest decrease
evidence would certainly not allow for any claim of of total and/or abdominal fat mass (Gruenewald and
established favorable long-term effects, there appears Matsumoto 2003). In a meta-analysis, testosterone
to be no basis to suggest that androgen therapy treatment was found to reduce fat mass by 1.6 kg
aiming at near physiological serum levels should be (95% CI: 2.5 to 0.6), corresponding to 6.2% (9.2 to
withheld out of fear of adverse effects on cardiovas- 3.3). Nevertheless, in this context GH may have a
cular risk (see Liu et al. 2003; Wu and von Eckard- larger effect than testosterone (Mnzer et al. 2001).
stein 2003; Kaufman and Vermeulen 2005). A recent The age-associated loss of muscle mass is accom-
report of increased incidence of cardiovascular- panied by decreased muscle strength, which occurs

348
Chapter 16: Androgens in male senescence

regardless of the level of physical activity (Rogers and (2002) observed improved leg and arm muscle
Evans 1993). Muscle weakness is an important com- strength and an increase in muscle net protein balance
ponent of frailty in old age, contributing to functional in a small number of older men treated with testoster-
limitation in activities of daily living and related prob- one for six months. Muscle strength measured by
lems such as an increased risk for falls (Rubenstein isokinetic peak torque was increased in flexion of the
et al. 1994; Guralnik et al. 1995; Bhasin and Tenover dominant knee, but not in knee extension or shoulder
1997; Rolland et al. 2011). Information on the associ- contraction during three months of transdermal
ation of endogenous androgens and muscle mass is administration of DHT in older men with low-normal
scarce. Van den Beld et al. (2000) and Vermeulen et al. serum testosterone (Ly et al. 2001). In this last study
(1999a) found no association of serum (free or non there was no effect of treatment on tests of gait, balance
SHBG-bound) testosterone with lean body mass in or mobility; no effect of treatment on muscle strength
sizable populations of ambulant elderly men. Simi- was observed in the studies by Kenny et al. (2001) and
larly, Roy et al. (2002) found no association of lean Snyder et al. (1999a). Crawford et al. (2003) reported
mass with testosterone, independent of age, in men increased muscle mass and strength during androgen
aged 20 to 90 years from the Baltimore Longitudinal treatment in men with mean age around 60 years
Study of Aging. Several studies suggest a positive asso- under treatment with glucocorticoids. In intermedi-
ciation of serum (free or bioavailable) testosterone ate-frail and frail elderly men, six months of testoster-
levels with muscle function and strength (Abbasi one treatment had beneficial effects on body
et al. 1993; Hkkinen and Pakarinen 1994; van den composition, muscle strength, physical function and
Beld et al. 2000; Roy et al. 2002). Recent, large, cross- quality of life (Srinivas-Shankar 2010); beneficial
sectional and longitudinal observational studies have effects were not maintained six months after end of
shown associations of low serum (free or bioavailable) treatment (OConnell et al. 2011). In elderly men with
testosterone with prevalent as well as worsening frailty limited mobility and high prevalence of chronic dis-
in elderly men (Orwoll et al. 2006b; Cawthon et al. ease, transdermal testosterone treatment planned for
2009; Hyde et al. 2010; Krasnoff et al. 2010; Travison six months improved muscle strength and stair
et al. 2011), although this was not confirmed in all climbing, but the trial was prematurely discontinued
studies (Mohr et al. 2007; Schaap et al. 2008). because of a high rate of adverse events (Basaria et al.
Experimental exposure of elderly men to testoster- 2010). In a study in older men with low-normal serum
one levels varying from hypogonadal to supraphysio- testosterone, Emmelot-Vonk et al. (2008) observed an
logical serum levels resulted in dose-dependent increase of lean body mass, but no beneficial effect on
increases of muscle mass and strength (Bhasin et al. functional status and mixed metabolic effects of six
2005; Sattler et al. 2009). The response to testosterone months testosterone therapy.
in the elderly appears similar to that in young men The data discussed in this section indicate that an
(Bhasin et al. 2005). Several controlled studies of age-associated partial hypoandrogenism might play a
more than three months duration with androgen contributory role in the sarcopenia of older men,
administration in elderly men with low or (low) which is multifactorially determined, and that the
normal serum testosterone (see Gruenewald and Mat- elderly can respond to pharmacological treatment
sumoto 2003; Isidori et al. 2005b for review) have with testosterone, in particular if serum testosterone
shown increases of lean body mass (Snyder et al. is initially low. However, whether androgen treatment
1999a; Kenny et al. 2001; Mnzer et al. 2001; Fer- plays a role in the management of sarcopenia and
rando et al. 2002; Steidle et al. 2003; Basaria et al. frailty in elderly men is not clear, as several issues
2010). Findings on muscle force and various meas- need to be clarified, such as optimal treatment modal-
ures of functionality have been heterogeneous (Isidori ities, safety in frail elderly, and longer-term persist-
et al. 2005b; Rolland et al. 2011 for review). Sih et al. ence of effects.
(1997) observed a significant increase in grip strength
in testosterone-treated men over the age of 50 years
(mean age 68 years) with low bioavailable serum 16.4.5 Senile osteoporosis
testosterone, in a prospective, randomized, placebo- Aging of men is accompanied by continuous bone
controlled trial of 12 months duration; lower extrem- loss, which persists and may even accelerate in old
ity muscle strength was not evaluated. Ferrando et al. age, and by exponential increase of vertebral and hip

349
Chapter 16: Androgens in male senescence

fracture incidence. Although age-specific fracture degree relatives, were found to be associated with a
incidence in men is only about half that in women, polymorphism of the CYP19 gene that encodes
one out of three to four fractures in the elderly occurs for the aromatase enzyme, independently of serum
in men, and the consequences of fractures in men are estradiol levels, suggesting indirectly that local aroma-
more severe in terms of morbidity and mortality (see tization of testosterone in bone tissue might play a
Orwoll and Klein 1995; Kaufman and Goemaere role (Van Pottelbergh et al. 2003a). Low estradiol
2008; Khosla 2010 for review). levels have been associated with prevalent vertebral
Profound hypogonadism in older men induces fractures in elderly men of the Rancho Bernardo
high bone turnover, accelerated bone loss and Study (Barrett-Connor et al. 2000). In the MrOS
increased fracture risk, similar to the effects seen in Sweden cohort, low serum estradiol and high serum
younger men (Stoch et al. 2001; Mittan et al. 2002; SHBG were associated with increased fracture risk
Shahinian et al. 2005; Fink et al. 2006), and there are (Mellstrm et al. 2008). There are indications for a
indications that TRT in men with acquired hypogo- serum (bioavailable) estradiol threshold effect for
nadism may result in partial recovery of bone density both bone loss and fracture risk (Mellstrm et al.
(Katznelson et al. 1996; Behre et al. 1997). Hypogo- 2008; Khosla 2010).
nadism has also been reported to be a risk factor for In community-dwelling men over age 70 years, we
hip fracture in elderly men (Stanley et al. 1991; Jack- found no association of BMD or bone metabolism
son et al. 1992; Boonen et al. 1997), but, besides low with a CAG-repeat polymorphism of the androgen
bone mass, other factors related to testosterone, such receptor gene (Van Pottelbergh et al. 2001), while such
as muscle weakness, may be involved in the occur- an association has been reported for younger men
rence of hip fractures. In community-dwelling men (Zitzmann et al. 2001; Guadalupe-Grau et al. 2010).
over the age of 60 years from the Dubbo Osteoporosis Currently available, controlled data on the effects
Epidemiology Study, serum testosterone adjusted for of testosterone treatment in elderly men are limited.
serum SHBG and estradiol was associated with risk Observed effects of androgen treatment in elderly
for osteoporotic factures (Meier et al. 2008). men on biochemical indices of bone turnover
There is strong evidence that aromatization of have been rather heterogeneous (Orwoll and Oviatt
testosterone to estradiol is of major importance for 1992; Sih et al. 1997; Snyder et al. 1999b; Kenny et al.
preservation of the skeleton in elderly men. 2001), although in a meta-analysis treatment does
A preponderant role of estrogen in the regulation of appear to reduce levels of bone resorption markers
bone metabolism in elderly men has been elegantly (Isidori et al. 2005b).
demonstrated in a short-term intervention study with In a randomized, double-blind trial of transder-
selective manipulation of testosterone and estradiol mal administration of testosterone by scrotal patch
levels (Falahati-Nini et al. 2000). This is further con- (6 mg/day) or placebo to men with normal or low
firmed by bone loss observed in elderly men during serum testosterone (n = 108), Snyder et al. (1999b)
treatment with an aromatase inhibitor that increased found that, after three years of treatment, lumbar
testosterone and decreased estradiol levels (Burnett- spine bone mineral density was increased in the pla-
Bowie et al. 2009). In cross-sectional studies in elderly cebo group as well as in the active treatment group,
men, associations of bone mineral status or biochem- both receiving calcium and vitamin D supplements,
ical markers of bone turnover with sex steroid levels without significant testosterone treatment effect; there
have been rather weak, although statistically signifi- was no change in BMD at the hip region in either
cant. In several of the latter studies (free or bioavail- treatment group. It is interesting to note that these
able) estradiol was more strongly associated with negative findings for testosterone effects on bone were
BMD and/or bone turnover markers than (free or accompanied by significant treatment effects on fat
bioavailable) testosterone. Serum (free or bioavail- mass and lean body mass. Kenny et al. (2001)
able) estradiol, but not serum testosterone, is reported prevention of bone loss at the hip during
inversely associated with prospectively assessed bone one year of treatment with transdermal testosterone
loss in elderly men (Khosla et al. 2001; Van Pottel- (5 mg/day by body patch) as compared to placebo in
bergh et al. 2003a; Cauley et al. 2010). Moreover, healthy elderly men aged 65 to 87 years, but differ-
changes in BMD, the personal clinical fracture history ences between placebo and active treatment were
of the subjects and the fracture history in their first- rather small. In a study of elderly men with initially

350
Chapter 16: Androgens in male senescence

moderately low serum testosterone, Amory et al. testosterone and to effects on the lumbar spine. In
(2004) observed a significant increase of BMD at the view of the modest bone effects, of the lack of relevant
spine and hip compared to placebo during 36 months studies with testosterone specifically in osteoporotic
of treatment with testosterone enanthate im 200 mg/ men, and of the availability of better validated specific
wk, with or without associated treatment with the osteoporosis medication such as bisphosphonates,
5a-reductase inhibitor finasteride. In frail older men osteoporosis as such is not an indication for testoster-
with low bioavailable serum testosterone, a history of one therapy.
fracture and/or low BMD, Kenny et al. (2010)
observed an increase of spine BMD during transder-
mal testosterone treatment (5 mg/day), but interpret- 16.4.6 Additional clinical variables
ation is limited by a high drop-out rate. Crawford Men have higher hemoglobin levels and red cell mass
et al. (2003) found a beneficial effect of 12 months than women, and testosterone has a dose-dependent
administration of testosterone, but not of the only stimulatory effect on erythropoiesis that is more pro-
minimally aromatizable androgen nandrolone, on nounced in elderly compared to young men (Coviello
BMD in men treated with glucocorticoids who et al. 2008). Elderly men tend to have similar or only
had initially low or low-normal serum testosterone slightly lower hematocrit than young men. There is
levels. In a meta-analysis and systematic review, tes- no documentation of association between endogen-
tosterone treatment was found to favorably affect ous androgen levels and parameters of erythropoiesis
BMD at the spine, but not at the hip (Isidori et al. in elderly men.
2005b; Tracz et al. 2006). Aging is associated with a deterioration of mul-
Compared to findings of testosterone administra- tiple aspects of cognitive performance. Hypogona-
tion in younger hypogonadal men on bone effects, dal men tend to show diminished spatial skills and,
these are rather disappointing and may be explained albeit findings have not been consistent, several
by the fact that many men in these studies had either observational studies in elderly men have reported
normal or near normal initial androgen levels; a fur- positive associations of cognitive functions such as
ther explanation might be the possible existence of a verbal memory and spatial skills with endogenous
threshold effect with testosterone and derived estro- androgen levels (Kaufman and Vermeulen 2005 for
gen requirements for near maximal bone effects lying review). In a long-term follow-up study, low serum
at the lower end of the physiological range for sex testosterone was associated with increased risk of
steroid levels, which would be in line with the estra- Alzheimers disease (Moffat et al. 2004). In several,
diol threshold for bone loss and fracture risk reported but not all intervention studies, androgen adminis-
in several observational studies. A post-hoc analysis of tration to elderly men resulted in improved spatial
the study by Snyder et al. (1999b), indicating that only cognition and working memory, with decreased
those men with initially low serum total testosterone verbal fluency (see Muller et al. 2003; Gruenewald
(< 300350 ng/dl) increased their BMD during treat- and Matsumoto 2003 for review). In a study in men
ment supports this view, as do the positive results 66 to 86 years of age with normal cognitive func-
from the study by Amory et al. (2004) which included tion, treatment with testosterone enanthate 200 mg
men with generally lower serum testosterone every other week, a supraphysiological dose,
(i.e. baseline testosterone 283  49 ng/dl). resulted in a deterioration of cognitive functions
In summary, the evidence suggests that declining (Maki et al. 2007).
sex steroid levels may adversely impact on the preser- Endogenous free and bioavailable testosterone
vation of skeletal integrity in elderly men, and indi- levels were reported to be inversely associated with
cates that aromatization of testosterone to estradiol is depressive mood and depressive symptoms in aging
a major component of the regulation of bone homeo- men (Barrett-Connor et al. 1999; Delhez et al. 2003),
stasis in the elderly. The skeletal effects of testosterone but other studies failed to confirm such associations
deficiency are modulated by aromatase activity, which (TSjoen et al. 2005b; Kaufman and Vermeulen 2005
in turn is affected by factors such as adiposity and for review), or reported the opposite association of
heredity. Testosterone treatment has mostly only less depressive symptoms in elderly men with lower
modest beneficial effects on BMD in the elderly; serum testosterone (Perry et al. 2001). Similarly, the
mainly limited to men with initially low serum findings on association of low serum testosterone in

351
Chapter 16: Androgens in male senescence

the elderly with prevalence and incidence of depres- are rather more or less independent reflections of
sion have been inconsistent (Delhez et al. 2003; generally poorer health status.
Joshi et al. 2010).
Aging evidently is often associated with a decrease
of quality of life. The few available studies failed to 16.5 Androgen therapy in elderly men
establish a relation between endogenous androgen 16.5.1 Who should be considered
levels and quality of life (QoL), such as evaluated with
the SF-36 questionnaire (Dunbar et al. 2001; TSjoen for treatment?
et al. 2004). Whereas testosterone therapy has been The age-associated decrease in serum testosterone
reported to improve some aspects of QoL in some levels raises the issue of the potential role of androgen
studies (Snyder et al. 1999a; Crawford et al. 2003; substitution in elderly males. This implies a number
Srinivas-Shankar et al. 2010), other studies failed to of questions which, with the currently available evi-
objectivate a beneficial effect of testosterone treat- dence, can at best receive pragmatic interim answers:
ment on overall QoL or mood in elderly men (Ly who should be treated, how and for how long?
et al. 2001; Kunelius et al. 2002; Gruenewald and As to the first question, in theory androgen
Matsumoto 2003; Steidle et al. 2003; Kaufman and administration to elderly men could be either substi-
Vermeulen 2005 for review). tutive to alleviate symptoms and prevent complica-
tions of partial or more complete androgen
deficiency, or rather pharmacological, to elderly
16.4.7 Conclusions men who are not necessarily androgen deficient, with
The extent of the clinical consequences of relative specific treatment goals such as the prevention or
hypoandrogenism in elderly men remains difficult to treatment of osteoporosis, of frailty, or adjuvant treat-
pinpoint. Nevertheless, in many instances there is a ment of ED. A few usually small-scaled studies per-
significant, albeit usually weak, association between taining to particular clinical situations with
some aging-related clinical changes and serum levels indications of potential treatment benefits and risks
of testosterone and/or its aromatized metabolite (Gruenewald and Matsumoto 2003; Kaufman and
estradiol, which generally persists after adjustment Vermeulen 2005; Fernandez-Balsells et al. 2010 for
for major confounders such as age and adiposity. In review) notwithstanding, for no single indication in
any case, it is also evident that the androgen status is elderly men is the present level of evidence sufficient
at best only one of many factors that influence the to justify pharmacological androgen treatment in
pace and clinical expression of the aging process in elderly men, thus leaving us with only the substitu-
men. Although associations between plasma andro- tive treatment to be considered at this time.
gen levels and the prevailing clinical status have been It is generally not a matter for debate that pro-
generally weak or even absent, this does not necessar- longed androgen deficiency in young men results in
ily imply that the decline in testosterone levels in symptoms affecting QoL and carries a risk for longer-
elderly men is not clinically relevant. Indeed, one term complications, so that intervention to reestablish
may question whether a single time-point androgen physiological androgen levels is required unless there
measurement for estimation of the androgen status is is a specific contraindication for such an intervention.
sufficiently representative of this status in preceding As to the elderly, there is no a priori medical or moral
years, during which clinical changes have accumu- justification for withholding the benefits of substitu-
lated to produce the present clinical picture in the tive treatment from symptomatic hypogonadal eld-
investigated subject. Moreover, serum levels are at erly men. Nevertheless a prudent, sufficiently
best imperfect parameters of testosterone action in conservative approach to such treatment seems advis-
the tissues. In this regard more recent prospective able in view of limited data and clinical experience for
studies tend to reveal more robust associations this population, the low specificity of the symptoms
between sex steroid levels and clinical parameters, of hypogonadism in the elderly, and the potential for
including clinically relevant end-points such as frailty, greater susceptibility for adverse treatment effects.
fractures or mortality. A crucial question remains This brings up the key issue of how to diagnose
whether low androgen status and clinical outcome androgen deficiency in elderly men and thus the
are causally and specifically related, or whether both question of what the testosterone requirements are

352
Chapter 16: Androgens in male senescence

in elderly men (Vermeulen 2001). The question to be We found that no more than 1% of healthy men
asked is whether elderly men are equally, less or more aged 2040 years old, but over 20% of men older than
sensitive to testosterone action compared to the 65 years, present with a total serum testosterone
young. The answer to this question is limited on the below 11 nmol/l (Fig. 16.2): a prevalence of low tes-
one hand by lack of specificity of signs and symptoms tosterone in the same order of magnitude as reported
of androgen deficiency, while on the other hand no by others for men over 65 years of age (Orwoll et al.
useful direct biochemical measure of tissue activity of 2006a; Araujo et al. 2007). Parameters of the biologic-
testosterone and its bioactive metabolites is available. ally active fraction of serum testosterone, i.e. serum
Moreover, adding to the complexity, testosterone free testosterone and bioavailable testosterone, are in
requirements for normal functioning may differ principle more appropriate for the evaluation of the
according to the tissue and physiological functions androgen status. Their use will result in classification
under consideration (Zitzmann et al. 2006), whereas of an even larger proportion of elderly men as being
the large between-subject variability of serum testos- hypoandrogenic, as the age-dependent decrease is
terone levels seen at all ages may be the expression of steeper than for total testosterone. On the other hand,
individual differences in androgen sensitivity (Crabbe in a number of situations with low serum SHBG, such
et al. 2007; Huhtaniemi et al. 2009). as in obesity and during treatment with glucocorti-
In view of the still unresolved issue of what should coids, these measurements may reveal a better pre-
be considered physiologic or optimal androgen served androgen status than indicated by serum total
levels in elderly men, in many cases a margin of testosterone. In any case, taking into account the
uncertainty will persist. Therefore diagnosis of hypo- present lack of information on the long-term
gonadism in elderly men should be based on an benefitrisk ratio for androgen treatment in elderly
appropriately conservative evaluation of clinical and men, and the uncertainty about androgen require-
hormonal findings (Kaufman and Vermeulen 2005; ments in the elderly, it seems advisable to apply the
Wang et al. 2008; Bhasin et al. 2011). above cutoff values conservatively. In such a conser-
As to the testosterone threshold levels compatible vative approach to treatment of the elderly, interven-
with a diagnosis of hypogonadism, in the absence of tion thresholds may differ from diagnostic criteria,
definitive evidence of altered sensitivity to androgens considering for treatment only those men with
in the elderly, the least arbitrary attitude is probably frankly low testosterone, i.e. a (free) testosterone level
to use the same lower normal limit as in young men, clearly low relative to the proposed diagnostic thresh-
i.e. around 11 nmol/l (or 320 ng/dl) for total serum olds for hypogonadism.
testosterone, around 0.225 nmol/l (or 6.5 ng/dl Available evidence does not support the alterna-
for serum free testosterone, and around 5 nmol/l (or tive view that treatment might be justified in men
145 ng/dl) for bioavailable (i.e. non-specifically with higher prevalent serum testosterone, i.e. within
bound) testosterone. Although values for the lower the lower normal range for young men, on the basis
end of the reference range for total testosterone in that elderly men would be less sensitive to androgen
young men reported by various, experienced action. Indeed, whereas reports of decreased tissue
groups are usually not very different, there is some concentration of androgen receptors in the elderly
variation, with proposed lower normal values usually (Roehrborn et al. 1987) might suggest decreased sen-
ranging between 280 and 350 ng/dl (9.7 and 12 nmol/ sitivity, data from functional studies indicate that in
l), and with differences explained by issues of meth- older men there is rather an increased sensitivity of
odology and standardization of testosterone assays LH secretion to the negative feedback action of tes-
and selection of the reference population (Rosner tosterone (Winters et al. 1984; 1997; Deslypere et al.
et al. 2007; Wang et al. 2008; Wheeler and Barnes 1987). In elegant studies with suppression of
2008; Bhasin et al. 2011). Using liquid chromatog- endogenous testosterone secretion and subsequent
raphytandem mass spectrometry (LC-MS/MS) to exposure to graded doses of exogenous testosterone,
measure serum testosterone in a selected group of Bhasin et al. (2005) showed that the elderly are as
healthy non-obese young men, Bhasin et al. (2011) responsive as the young to the anabolic effects of tes-
reported lower normal levels of 348 ng/dl and 7.0 ng/ tosterone, such as on fat mass, muscle mass and force,
dl for total and free testosterone, defined as the with indications of higher sensitivity to stimulation of
2.5th percentile. erythropoiesis and decrease of HDL-C. Furthermore,

353
Chapter 16: Androgens in male senescence

the lower level of the serum testosterone range in instruments to quantify symptoms of LOH and to
young men appeared to be a valid threshold for screen for androgen deficiency in elderly men (Hei-
occurrence of symptoms of hypogonadism also in nemann et al. 1999; Morley et al. 2000; Smith et al.
the elderly (Wu et al. 2010; Bhasin et al. 2011). More- 2000). These instruments lack specificity (Morley
over, as discussed above, whereas most studies on et al. 2000; Dunbar et al. 2001; Delhez et al. 2003;
testosterone administration to elderly men have TSjoen et al. 2004), and their use for screening or
included a large proportion of men with serum tes- diagnostic purposes should be discouraged (Wang
tosterone within the lower normal range for young et al. 2008; Bhasin et al. 2011). Indeed, considering
men, probably based on the rationale that these low- the high prevalence in older men of non-specific
normal levels might in fact be suboptimal for many of symptoms loosely associated with hypoandrogenism,
these subjects, treatment effects were generally disap- spontaneous active reporting of complaints may have
pointing for those men without clearly low initial the merit of higher specificity, whereas soliciting of
serum testosterone levels. complaints with screening questionnaires may lead to
As to serum gonadotropin levels, although a mark- over-diagnosis and overtreatment.
edly elevated serum LH certainly adds weight to the In conclusion, according to the present state of the
finding of a decreased serum testosterone, pointing art, androgen supplementation should probably only
towards a predominantly testicular factor in this par- be considered in the presence of androgen serum
ticular patient (Tajar et al. 2010), elevated serum LH is levels clearly below the lower normal limit for young
not a prerequisite for the diagnosis of testosterone men, together with unequivocal signs and symptoms
deficiency in older men, the age-associated decline of of androgen deficiency, after having excluded revers-
Leydig cell function being usually of mixed testicular ible causes of low serum androgen and after careful
and neuroendocrine origin, in particular whenever screening for contraindications. The decision to treat
overweight and/or other co-morbidities contribute to will finally depend upon the balance between possible
the low testosterone of the subject. benefits and risks, integrating the most recent evi-
As to objective signs of relative androgen defi- dence available for androgen treatment as well as the
ciency, a decrease of muscle mass and strength and evidence available for possible pharmacological and
a concomitant increase in central body fat and osteo- non-pharmacological alternatives. Indeed, the lack of
porosis can most easily be objectified, but are not reliable data on the long-term riskbenefit ratio
specific signs. Decreased libido and sexual desire, loss imposes a critical and conservative attitude in accord-
of memory, difficulty in concentration, forgetfulness, ance with a basic principle of clinical practice; i.e.
insomnia, irritability and depressed mood, as well as primum non nocere.
decreased sense of well-being, are rather subjective From a practical point of view, serum testosterone
feelings or impressions, less easily objectified and should be assessed in the morning before 10 a.m., and
not easy to differentiate from hormone-independent low values obtained when sampling has been per-
aging. Of the latter indications, sexual symptoms formed later during the day need to be verified by a
have been reported to be most consistently related to morning sample. Low values should be confirmed by
low serum testosterone (Wu et al. 2010). Complaints a second sample, ideally obtained with an interval of
of excessive sweating are not uncommon, whereas at least a couple of weeks. The reference range for
true hot flushes do occur in elderly men, although serum testosterone should be validated for the type of
they are mainly prevalent in severe acquired hypogo- assay used and preferably also for each laboratory;
nadism such as under hormonal treatment for pro- whereas for estimates of free or bioavailable testoster-
state cancer. one one should use valid methods. As to the latter, it
A number of questionnaires have been developed has been shown that free testosterone can be reliably
for use in clinical or epidemiological settings to help calculated from serum total testosterone and SHBG
describe and semi-quantify symptoms in different using an equation derived from the mass action law,
areas of relevance to elderly men, such as question- except when there are gross abnormalities in plasma
naires on self-perceived health status, on depressive proteins or when the subjects are treated with sub-
mood, on urinary symptoms, on erectile function, or stances with high affinity for SHBG (e.g. DHT), the
on coping with activities of daily living. There are also calculation being easily done with a simple algorithm
questionnaires that have been proposed as dedicated on a personal or pocket computer. Neither the

354
Chapter 16: Androgens in male senescence

commercially available direct immunoassays for free pharmacological doses, as used by bodybuilders.
testosterone, nor the simple ratio of total serum tes- Traditionally, it has been a matter of concern that
tosterone over serum SHBG should be regarded as prolonged treatment with androgen may increase
reliable options in men (Vermeulen et al. 1999b). the risk of cardiovascular disease. The complex rela-
tionship between endogenous and exogenous andro-
16.5.2 Potential benefits gens and cardiovascular risk is discussed in Section
16.4.3. In-depth review of the literature (Liu et al.
From the preceding sections of this chapter and from
2003; Wu and von Eckardstein 2003) suggests that
literature reviews (Gruenewald and Matsumoto 2003;
exogenous androgen administration has both poten-
Kaufman and Vermeulen 2005; Isidori et al. 2005a;
tially favorable as well as possible adverse effects on
2005b; Ottenbacher et al. 2006; Bhasin et al. 2010), it
cardiovascular risk. With the evidence currently avail-
emerges that testosterone administration to elderly
able, improving cardiovascular risk can certainly not
men can induce potentially beneficial effects, but the
be considered an indication for androgen treatment,
results are often mitigated and without demonstrated
but there is also no suggestion that treatment with
impact on end-points that are directly relevant for the
moderate, close to physiological doses carries an
clinic. Several studies have shown improvement of
unacceptable risk that should prevent initiating other-
lean body mass and sometimes also of muscle
wise indicated androgen treatment. A recent meta-
strength, but whether these changes are sufficient to
analysis (Fernandez-Balsells et al. 2010) found no
make a meaningful difference in terms of functional-
difference in the rates of death, myocardial infarction,
ity is still unclear. Positive effects on BMD are seen
revascularization procedures or cardiac arrhythmias
only in men with frankly low serum testosterone, and
between testosterone therapy and placebo/non-
we have no information on the effect of treatment on
intervention in adult men. Testosterone treatment is
fracture rates. Abdominal fat may decrease and insu-
associated with a decrease of HDL-C; the other lipid
lin sensitivity may improve, whereas high-dose testos-
fractions, blood pressure and incidence of diabetes are
terone may have direct beneficial effects on heart and
not affected.
arteries, but we have no indication of gains in terms
Androgen treatment results in a significant
of hard cardiovascular end-points. There have been
increase of hematocrit and blood hemoglobin level,
reports of favorable effects on mood, cognition and
and androgen treatment is associated with increased
general well-being, but the findings are not always
incidence of erythrocytosis (Calof et al. 2005; Fernan-
consistent and we have no data indicating that treat-
dez-Balsells et al. 2010). Moreover, the stimulatory
ment may prevent or help treat depression, or have
effects of testosterone administration on erythropoi-
substantial longer-term effects on cognition and QoL.
esis have been shown to be increased in the elderly as
Androgen therapy has beneficial effects on sexual
compared to young men (Coviello et al. 2008).
functioning in men with initially low serum testoster-
Polycythemia is not uncommon and may necessitate
one levels, although the treatment effects may be
dose reduction, temporary interruption of treatment,
small and usually significant for only some indices
or alternative measures such as phlebotomy. The
of sexual function.
occurrence of polycythemia appears to be more likely
A major limitation is the scarcity of controlled
when the subjects are exposed to markedly supraphy-
data. No more than a few hundred elderly men in
siological androgen levels, as is often the case with
total have been included in controlled trials, and
treatment regimens consisting of im administration
among these only a minority in trials of at least one
of depot preparations of testosterone esters at inter-
years duration; less than half these men received
vals of two to three weeks (Dobs et al. 1999). Never-
active treatment with testosterone. Moreover, a sub-
theless, significant increases of hematocrit and
stantial proportion of men included had initially
hemoglobin levels are also seen during transdermal
(low) normal serum testosterone.
administration of either testosterone (Snyder et al.
1999a; Steidle et al. 2003) or DHT (Ly et al. 2001;
16.5.3 Potential risks Kunelius et al. 2002), occasionally leading to erythro-
As to the risks of androgen replacement therapy in cytosis. Monitoring for occurrence of exaggerated
elderly men, we consider here only effects of physio- elevations of hematocrit or hemoglobin concentra-
logical doses of testosterone and not those of massive tions during androgen treatment in elderly men is

355
Chapter 16: Androgens in male senescence

advisable, keeping in mind that some patients, in Matsumoto 2003), and only in cases of severe lower
particular some with pulmonary disease, can have a urinary tract obstructive symptoms is benign prostate
high a priori risk of erythrocytosis. disease considered a contraindication for androgen
The assertion that testosterone therapy may exacer- treatment (Bhasin et al. 2003; 2010; Wang et al.
bate obstructive sleep apnea (Sandblom et al. 1983) is 2008). Prostate-specific antigen (PSA) levels, a par-
essentially based on a few case reports. Whereas largely ameter of androgen stimulation of prostate tissue,
supraphysiological testosterone doses may indeed increase moderately during treatment but usually
increase the risk of sleep apnea, this does not seem to within the normal range, and after stopping treatment
be the case during treatment with more physiological the values return to pretreatment levels (Behre et al.
dosages (Calof et al. 2005; Liu et al. 2007b). Neverthe- 1994, Hajjar et al. 1997). Small significant increases in
less, clinical practice guidelines suggest that testosterone PSA have also been observed under transdermal
therapy should not be initiated in men with severe administration of testosterone to elderly men; only
untreated sleep apnea (Bhasin et al. 2010). rarely is a more important rise in PSA seen (Snyder
Gynecomastia, related to the conversion of testos- et al. 1999a; Kenny et al. 2001). Levels of PSA and
terone to estradiol in peripheral tissues, mainly fat prostate size were reported to remain unchanged (Ly
tissue, which is relatively increased in elderly men, is et al. 2001; Kunelius et al. 2002) or even decreased (de
a not uncommon but benign side-effect in elderly Lignieres 1993) under transdermal DHT administra-
men, especially in the obese. This side-effect is prob- tion to older men, even though on close examination
ably less frequent when patients are not exposed to small PSA increases in comparison with placebo
supraphysiological serum levels of testosterone. might occur (Ly et al. 2001). One could hypothesize
Although testosterone causes some sodium and water that DHT treatment could have a relative prostate-
retention, this effect does usually not cause a problem, sparing effect through a reduction of estradiol and/or
although this might be relevant in patients with con- DHT levels in prostate tissue as a result of suppres-
gestive heart failure, hypertension or renal sion of concentrations in the systemic circulation of
insufficiency. Hepatotoxicity is very rare when non- their precursor, i.e. testosterone.
oral routes of administration of testosterone are used. Undoubtedly clinical prostate carcinoma is an
Of greater concern are the possible effects on the androgen-sensitive tumor. Hence presence of a clin-
prostate, which is an androgen-dependent organ ical prostate carcinoma is an absolute contraindica-
(Bhasin et al. 2003). In a meta-analysis by Calof tion to testosterone supplementation. Subclinical
et al. (2005), the combined rate of all prostate events carcinoma, only detectable on histology but undetect-
was significantly greater in testosterone-treated com- able by biochemical or clinical procedures, is found in
pared to placebo-treated men (odds ratio 1.78; 95% a majority of men over 70 years old. Only a small
CI: 1.072.95). Rates of prostate cancers, PSA greater minority of these subclinical carcinomas will further
than 4 ng/dl and prostate biopsies were numerically, develop to a clinical carcinoma. It is not known
but not significantly greater in testosterone-treated whether testosterone treatment would stimulate the
men. In a recent meta-analysis, Fernandez-Balsells progression of subclinical carcinoma, and so far no
et al. (2010) found no significant effect of testosterone available data indicate that testosterone substitution
therapy on outcomes such as prostate cancer inci- will activate subclinical carcinoma (Jackson et al.
dence or need for prostate biopsy when compared to 1989; Schrder 1996). However, all studies so far
placebo/non-intervention. concern only small numbers of carefully selected eld-
Concerning benign prostatic hyperplasia (BPH), erly males treated for short periods of time. In any
studies to date failed to observe important growth of case, before starting testosterone supplementation
the prostate (Wallace et al. 1993; Behre et al. 1994) careful exclusion of a prostate carcinoma by rectal
and failed to find any relationship between plasma examination and serum PSA and, when required,
and BPH tissue levels of testosterone, DHT or estra- supplemented by ultrasonography and biopsy, is
diol. It appears that tissue levels are determined by mandatory. It is advised to perform controls of rectal
enzyme activity in the tissue itself, rather than by examination, PSA and a symptom questionnaire for
surrounding plasma androgen levels. Treatment does benign prostatic hyperplasia after 3 to 6 and 12
not seem to result in increased voiding symptoms or months in stable patients; thereafter yearly controls
postvoid residual volume (Gruenewald and (Bhasin et al. 2003; 2010; Wang et al. 2008). Referral

356
Chapter 16: Androgens in male senescence

for urological work-up prior to therapy is required in ongoing research aimed at the development of select-
case of palpable nodule or indurations on rectal ive androgen receptor modulators having tissue-
examination, PSA level above 4 ng/ml or above 3 ng/ specific properties. Moreover, other options such as
ml in subjects at higher risk for prostate cancer. stimulation of endogenous sex steroid production
During therapy, a palpable nodule or indurations, and a possible role for selective estrogen receptor
an increase of PSA greater than 1.4 ng/dl or a PSA modulators for particular indications in elderly
velocity of more than 0.4 ng/mlyr using six-month men (e.g. osteoporosis) might also deserve to be
PSA as reference, or an International Prostate Symp- further explored. With the currently available
tom Score (IPSS) greater than 19 should also prompt evidence there is no place for treatment with DHEA
further urological evaluation (Bhasin et al. 2010). in elderly men.
For testosterone administration, new improved
16.5.4 Modalities of androgen options have become available that avoid the large
periodic oscillations between markedly supraphysio-
substitution logical testosterone levels and deficiently low con-
In young healthy men, testosterone levels vary centrations, and some of these newer modes of
between 11 and 35 nmol/l and show a circadian administration allow individual tailoring of treat-
variation with amplitude of  35%, highest levels ment. The pharmacology and practical aspects of
being reached in the early morning and nadir values testosterone replacement are discussed in detail in
in the evening around 68 p.m. Therefore, when Chapter 13.
supplementing testosterone, the aim should be to
alleviate symptoms related to the relative androgen
deficiency, if possible by achieving plasma testoster- 16.6 Key messages
one levels in the physiological range. As discussed in
 Mean total serum testosterone decreases
Section 16.5.1, in the absence of convincing evidence progressively in healthy men over the age of
that androgen requirements change with age, physio- 60 years (30% decrease between the ages of 25
logical levels in young men should be aimed for. and 75 years). Age-associated decrease of the
There is presently no evidence that it is clinically bioavailable fractions of serum testosterone is
important to mimic the nycthemeral variations as steeper as a consequence of an age-related
found in young adults. Nevertheless, constant levels increase of serum SHBG (50% decrease of free
in the upper normal range will result in 24-hour mean or bioavailable testosterone between the ages of
levels that are supraphysiological as compared to the 25 and 75 years).
situation in young men subject to nycthemeral vari-  There is great interindividual variability of
prevailing androgen levels in the elderly,
ations of serum testosterone.
ranging from perfectly preserved to frankly
Taken that the hypothalamic-pituitary-testicular
hypogonadal. Part of the interindividual
axis is sensitive to negative feedback, and even more so variability in serum testosterone levels is
in elderly males, the dose administered should increase explained by heredity, physiological factors and
testosterone levels up to the physiological range and not lifestyle-related factors.
merely suppress LH secretion with only replacement of  The proportion of men with subnormal
the deficient testosterone production by an inadequate testosterone relative to levels in young men
dose of exogenous testosterone. In practical terms, full increases with age (>20% after age 60 years);
replacement doses are usually required. whether androgen requirements change in
Obviously, considering the fact that metabolization aging men remains to be definitely
of testosterone to DHT and estradiol constitutes an established, but available evidence
suggests that androgen requirements are
important component for the regulation and full
not increased.
expression of testosterone effects, treatment with tes-
 The age-related decline in Leydig cell function
tosterone is the most physiological approach and the can be accentuated transiently or more
preferable option with the currently available evidence, permanently by co-morbidity and medication.
but the debate is certainly not closed in view of inter-  The contribution of lifestyle-related factors and/
esting data obtained with alternative treatments such or subclinical co-morbidities in the age-related
as transdermal DHT and in view of the intensive

357
Chapter 16: Androgens in male senescence

decline of serum testosterone in apparently of androgen deficiency, in the absence of other


healthy men remains an area of debate. reversible causes of decreased androgen levels
 The age-related decline of testosterone and after screening for contraindications.
production is the result of primary testicular  Available dedicated questionnaires assessing
changes as well as of failure of the hypothalamic aging male symptomatology lack specificity, and
regulatory mechanisms to respond adequately to their use for screening purposes should be
the hypoandrogenic state. discouraged.
 Many of the clinical features of aging in men are  The longer-term riskbenefit ratio for androgen
reminiscent of clinical changes seen in administration to elderly men is unknown.
hypogonadism in younger men; relative  Possible benefits of the treatment include an
hypoandrogenism may be involved in some, but improved sense of general well-being, of
certainly not all clinical changes. libido and of muscle strength, with increase
 Although testosterone levels required for normal of lean body mass and limited decrease of fat
sexual activity may be rather low and the factors mass.
most commonly involved in sexual dysfunction in  So far, the limited data on safety of TRT in the
elderly men are not hormonal, serum elderly have been rather reassuring: larger-scale
testosterone levels below the range for young studies of longer duration are still needed to
men are associated with sexual symptoms. assess safety, in particular at the prostate level;
 Hypoandrogenism may be involved in the development of erythrocytosis is one of the most
sarcopenia of elderly men. troublesome side-effects, which may be less
 The role of hypoandrogenism in male senile frequent if largely supraphysiological androgen
osteoporosis remains to be confirmed; recent levels are avoided.
data indicate that aromatization of  Androgen replacement therapy in the elderly
testosterone to estradiol plays an important role requires careful monitoring by an experienced
in the regulation of bone metabolism in elderly physician.
men.
 In the present state of the art, androgen
supplementation should only be considered in
the presence of androgen serum levels clearly
Acknowledgement
below the lower normal limit for younger men, Part of this work was supported by the Fonds voor
together with unequivocal signs and symptoms Wetenschappelijk Onderzoek Vlaanderen, Grants
G.3062.92 and G.0058.97 and G.0331.02.

16.7 References testosterone. J Clin Endocrinol


Metab 89:503510
in men. J Clin Endocrinol Metab
92:42414247
Abbasi AA, Drinka PJ, Mattson DE,
Rudman D (1993) Low circulating Andersson AM, Jensen TK, Juul A, Araujo AB, Dixon JM, Suarez EA,
levels of insulin-like growth Petersen JH, Jrgensen, Skakkebaek Murad MH, Guey LT, Wittert GA
factors and testosterone in NE (2007) Secular decline in male (2011) Clinical review: endogenous
chronically institutionalized testosterone and sex hormone testosterone and mortality in men: a
elderly men. J Am Geriatr Soc binding globulin serum levels in systematic review and meta-
41:975982 Danish population surveys. analysis. J Clin Endocrinol Metab
J Clin Endocrinol Metab 96:30073019
Alexandersen P, Haarbo J, Christiansen 92:46964705
C (1996) The relationship of natural Aversa A, Isidori AM, Spera G,
Andersson B, Marin P, Lissner L, Lenzi A, Fabbri A (2003)
androgens to coronary heart disease
Vermeulen A, Bjrntorp P Androgens improve cavernous
in males: a review. Atherosclerosis
(1994) Testosterone
125:113 vasodilation and response to
concentration in women and men sildenafil in patients with erectile
Amory JK, Watts NB, Easley KA, with NIDDM. Diabetes Care dysfunction. Clin Endocrinol
Sutton PR, Anawalt BD, Matsumoto 17:405411 58:632638
AM, Bremner WJ, Tenover JL Araujo AB, Esche GR, Kupelian V,
(2004) Exogenous testosterone or Bagatell CJ, Bremner WJ (1995)
ODonnell AB, Travison TG,
testosterone with finasteride Androgen and progestogen effects
Williams RE, Clark RV, McKinlay
increases bone mineral density in on plasma lipids. Prog Cardiovasc
JB (2007) Prevalence of
older men with low serum Dis 38:255271
symptomatic androgen deficiency

358
Chapter 16: Androgens in male senescence

Bagatell CJ, Knopp RH, Vale WW, Appleman E, Aggarwal S, Bhasin G, Dzekov C, Dzeko v J, Magliano L,
Rivier JE, Bremner WJ (1992) Hede-Brierley L, Bhatia A, Collins Storer TW (2005) Older men are as
Physiologic testosterone levels in L, LeBrasseur N, Fiore LD, Bhasin S responsive as young men to the
normal men suppress high-density (2010) Adverse events anabolic effects of graded doses
lipoprotein cholesterol levels. Ann associated with testosterone of testosterone on the skeletal
Int Med 116:967973 administration. N Engl J Med muscle. J Clin Endocrinol Metab
Baker HWG, Burger HG, de Kretser D, 363:109122 90:678688
Hudson B (1977) Endocrinology of Behre HM, Bohmeyer J, Nieschlag E Bhasin S, Cunningham GR, Hayes FJ,
aging: pituitary testicular axis. In: (1994) Prostate volume in Matsumoto AM, Snyder PJ,
James VHT (ed) Proceedings of the testosterone-treated and untreated Swerdloff RS, Montori VM (2010)
5th International Congress of hypogonadal men in comparison to Testosterone therapy in men
Endocrinology. Excerpta Medica age-matched normal controls. Clin with androgen deficiency
Foundation, Amsterdam, Endocrinol 40:341349 syndromes: an Endocrine
pp 479183 Society Clinical Practice Guideline.
Behre HM, Kliesch S, Leifke E, Link
Baker HWG, Burger HG, de Kretser J Clin Endocrinol Metab 95:
TM, Nieschlag E (1997) Long term
DM, Dulmanis A, Hudson B, 25362559
effect of testosterone therapy on
OConnor S, Paulson CA, Purcell N, bone mineral density in Bhasin S, Pencina M, Jasuja GK,
Rennie GC, Seah CS, Taft HP, hypogonadal men. J Clin Endocrinol Travison TG, Coviello A, Orwoll E,
Wang C (1979) A study of the Metab 82:23862390 Wang PY, Nielson C, Wu F, Tajar
endocrine manifestations of hepatic A, Labrie F, Vesper H, Zhang A,
Bergendahl M, Aloi JA, Iranmanesh A,
cirrhosis. Q J Med 177:145178 Ulloor J, Singh R, DAgostino R,
Mulligan TM, Veldhuis JD (1998)
Bancroft J (1984) Androgens, sexuality Vasan RS (2011) Reference ranges
Fasting suppresses pulsatile
and the aging male. In: Labrie F, for testosterone in men generated
luteinizing hormone (LH) secretion
Proulx L (eds) Endocrinology. using liquid chromatography
and enhances order lines of LH
Elsevier, Amsterdam, pp 913916 tandem mass spectrometry in a
release in young but not in older
community-based sample of healthy
Barrett-Connor E, Von Mhlen DG, men. J Clin Endocrinol Metab
nonobese young men in the
Kritz-Silverstein D (1999) 83:19671975
Framingham Heart Study and
Bioavailable testosterone and Bhasin S, Tenover JS (1997) Age- applied to three geographically
depressed mood in older men: the associated sarcopenia issues in the distinct cohorts. J Clin Endocrinol
Rancho Bernardo Study. J Clin use of testosterone as an anabolic Metab 96:24302439
Endocrinol Metab 84:573577 agent in older men. J Clin Bixler EO, Santen RJ, Kales A (1977)
Barrett-Connor E, Mueller JE, Von Endocrinol Metab 82:
Inverse effects of thioridazine
Mhlen DG, Laughlin GA, 16591660
(Melleril) on serum prolactin and
Schneider DL, Sartoris DJ (2000) Bhasin S, Woodhouse L, Casabury R, testosterone concentrations in
Low levels of estradiol are associated Singh AB, Bhasin D, Berman N, normal men. In: Troen P, Nankin
with vertebral fractures in older Chen XH, Yarasheski KE, Magliano HR (eds) The Testis in Normal
men: the Rancho Bernardo Study. L, Dzekov C, Dzekov J, Bross R, and Infertile Men. Raven, New York,
J Clin Endocrinol Metab 85:219223 Phillips J, Sinha-Hikim I, Shen RQ, pp 405409
Barrett-Connor EL, Khaw KT (1987) Storer TW (2001) Testosterone Bogaert V, Taes Y, Konings P, Van
Cigarette smoking and serum sex dose-response relationships in Steen K, De Bacquer D, Goemaere
hormones in men. Am J Epidemiol healthy young men. Am J Physiol S, Zmierczak H, Crabbe P, Kaufman
128:796805 Endocrinol Metab 281: E1172 JM (2008) Heritability of blood
E1181 concentrations of sex-steroids in
Basaria S, Lieb II J, Tang AM, DeWeese
T, Carducci M, Eisenberger M, Bhasin S, Singh AB, Mac RP, Carter B, relation to body composition in
Dobs AS (2002) Long-term effects Lee MI, Cunningham GR (2003) young adult male siblings. Clin
of androgen deprivation therapy in Managing the risk of prostate Endocrinol 69:129135
prostate cancer patients. Clin disease during testosterone Bogaert V, Vanbillemont G, Taes Y, De
Endocrinol 56:779786 replacement therapy in older men: Bacquer D, Deschepper E, Van
Basaria S, Coviello AD, Travison TG, recommendations for a Steen K, Kaufman JM (2009) Small
Storer TW, Farwell WR, Jette AM, standardized monitoring plan. effect of the androgen receptor gene
Eder R, Tennstedt S, Ulloor J, J Androl 24:299311 GGN repeat polymorphism on
Zhang A, Choong K, Lakshman Bhasin S, Woodhouse L, Casaburi R, serum testosterone levels in
KM, Mazer NA, Miciek R, Krasnoff Singh AB, Phong Mac R, Lee M, healthy men. Eur J Endocrinol
J, Elmi A, Knapp PE, Brooks B, Yarasheski KE, Sinha-Hikim I, 161:171177

359
Chapter 16: Androgens in male senescence

Boonen S, Vanderschueren D, Xiao Korach KS, Simpson ER (1997) men with gonadotropin suppression
GC, Verbeke G, Dequeker J, Effect of testosterone and estradiol receiving graded doses of
Geusens P, Broos P, Bouillon R in a man with aromatase deficiency. testosterone. J Clin Endocrinol
(1997) Age related (Type II) femoral N Engl J Med 337:9195 Metab 91:46694672
neck osteoporosis in men: Cauley JA, Ewing SK, Taylor BC, Fink Coviello AD, Kaplan B, Lakshman KM,
biochemical evidence for both HA, Ensrud KE, Bauer DC, Barrett- Chen T, Singh AB, Bhasin S (2008)
hypovitaminosis D- and androgen Connor E, Marshall, Orwoll ES, for Effects of graded doses of
deficiency-induced bone resorption. the Osteoporotic Fractures in Men testosterone on erythropoiesis in
J Bone Miner Res 12:21192126 Study (MrOS) Research Group healthy young and older men. J Clin
Brambilla DJ, ODonnell AB, (2010) Sex steroid hormones in Endocrinol Metab 93:914919
Matsumoto AM, McKinlay (2007) older men: longitudinal associations Crabbe P, Bogaert V, De Bacquer D,
Lack of seasonal variation in serum with 4.5-year change in hip bone Goemaere S, Zmierczak H,
sex hormone levels in middle-aged mineral density the osteoporotic Kaufman JM (2007) Part of the
to older men in the Boston area. fractures in men study. J Clin interindividual variation in serum
J Clin Endocrinol Metab 92: Endocrinol Metab 95:43144323 testosterone levels in healthy men
42244229
Cawthon PM, Ensrud KE, Laughlin reflects differences in androgen
Bremner WJ, Vitiello MV, Prinz PN GA, Cauley JA, Dam TTL, Barrett- sensitivity and feedback set point:
(1983) Loss of circadian rhythmicity Connor E, Fink HA, Hoffman AR, contribution of the androgen
in blood testosterone levels with Lau E, Lane NE, Stefanick ML, receptor polyglutamine tract
aging in normal men. J Clin Cummings SR, Orwoll ES, for the polymorphism. J Clin Endocrinol
Endocrinol Metab 56:12781281 Osteoporotic Fractures in Men 92:36043610
Brenta G, Schnitman M, Gurfinkiel M, (MrOS) Research Group (2009) Sex Crawford BAL, Liu PY, Kean MT,
Damilano S, Pierini A, Sinay I, hormones and frailty in older men: Bleasel JF, Handelsman DJ (2003)
Pisarev MA (1999) Variations of sex the Osteoporotic Fractures in Men Randomized placebo-controlled
hormone-binding globulin in (MrOS) Study. J Clin Endocrinol trial of androgen effects on muscle
thyroid dysfunction. Thyroid Metab 94:38063815 and bone in men requiring long-
9:273277 Cicero TJ (1982) Alcohol induced term systemic glucocorticoid
Bross R, Javanbakth M, Bhasin S (1999) defects in the hypothalamo- treatment. J Clin Endocrinol Metab
Anabolic interventions for aging- pituitary luteinizing hormone 88:31673176
associated sarcopenia. J Clin action in the male. Alcoholism Dai WS, Gutai JP, Kuller LH, Cauley
Endocrinol Metab 84:34203430 6:207215 JA (1988) Cigarette smoking and
Buena F, Swerdloff RS, Steiner BS, Cooper TG, Keck C, Oberdieck U, serum sex hormones in men. Am
Lutchmansingh P, Peterson MA, Nieschlag E (1993) Effects of J Epidemiol 128: 796805
Pandian MR, Galmarini M, Bhasin multiple ejaculations after extended Daniell HW (2002) Hypogonadism in
S (1993) Sexual function does not periods of sexual abstinence on total men consuming sustained-action
change when serum testosterone motile and normal sperm numbers oral opioids. J Pain 3:377384
levels are pharmacologically varied as well as on accessory gland
within the normal male range. Fertil secretions from healthy normal and Davidson JM, Chen JJ, Crapo L, Gray
Steril 59:11181123 oligospermic men. Hum Reprod GD, Greenleaf WJ, Catania JA
8:12511258 (1983) Hormonal changes and
Burnett-Bowie SA, McKay EA, Lee H,
sexual function in aging men. J Clin
Leder BZ (2009) Effects of Couillard C, Gagnon J, Bergeron J, Endocrinol Metab 57:7177
aromatase inhibition on bone Leon AS, Rao DC, Skinner JS,
mineral density and bone turnover Wilmore JH, Desprs JP, Bouchard Delhez M, Hansenne M, Legros J-J
in older men with low testosterone C (2000) Contribution of body (2003) Andropause and
levels. J Clin Endocrinol Metab fatness and adipose tissue psychopathology: minor symptoms
94:47854792 distribution to the age variation in rather than pathological ones.
Calof OM, Singh AB, Lee ML, Kenny plasma steroid hormone Psychoneuroendocrinology 28:
AM, Urban RJ, Tenover JL, Bhasin concentration in men: the 863874
S (2005) Adverse events associated HERITAGE family study. J Clin de Lignieres B (1993) Transdermal
with testosterone supplementation Endocrinol Metab 10261031 dihydrotestosterone treatment of
of older men. J Gerontol A Biol Sci Coviello AD, Lakshman K, Mazer NA, andropause. Ann Med 25:235241
Med Sci 60:14511457 Bhasin S (2006) Differences in the Deslypere JP, Vermeulen A (1981)
Carani C, Qin K, Simoni M, Faustini- apparent metabolic clearance rate of Aging and tissue androgens. J Clin
Fustini M, Serpente S, Boyd J, testosterone in young and older Endocrinol Metab 53:430434

360
Chapter 16: Androgens in male senescence

Deslypere JP, Vermeulen A (1985) Effect of testosterone Massachusetts Male Aging Study.
Influence of age on steroid supplementation on functional J Clin Endocrinol Metab 87:589598
concentration in skin and striated mobility, cognition, and other Fernandez-Balsells MM, Murad MH,
muscle in women and in cardiac parameters in older men. JAMA Lane M, Lampropulos JF,
muscle and lung tissue in men. 299:3952 Albuquerque F, Mullan RJ, Agrwal
J Clin Endocrinol Metab 60:648653 Erfurth EM, Hagmar LE, Sf M, Hall N, Elamin MB, Gallegos-Orozco JF,
Deslypere JP, Sayed A, Punjabi U, K (1996) Serum levels of insulin-like Wang AT, Erwin PJ, Bhasin S,
Verdonck L, Vermeulen A (1982) growth factor I and insulin-like Montori VM (2010) Adverse effects
Plasma 5a androstane-3a,17-diol growth factor binding protein 1 of testosterone therapy in adult
and urinary 5a-androstane, 3a17- correlate with serum free men: a systematic review and meta-
diol glucuronide, parameters of testosterone and sex hormone analysis. J Clin Endocrinol Metab
peripheral androgen action: a binding globulin in healthy young 95:25602575
comparative study. J Clin Endocrinol and middle-aged men. Clin Ferrando AA, Sheffield-Moore M,
Metab 54:386391 Endocrinol 44:659664 Yeckel CW, Gilkison C, Jiang J,
Deslypere JP, Kaufman JM, Eriksson AL, Lorentzon M, Mellstrm Achacosa A, Lieberman SA, Tipton
Vermeulen T, Vogelaers D, D, Vandenput L, Swanson C, K, Wolfe RR, Urban RJ (2002)
Vandalem JL, Vermeulen A (1987) Andersson N, Hammond GL, Testosterone administration to
Influence of age on pulsatile Jakobsson J, Rane A, Orwoll ES, older men improves muscle
luteinizing hormone release and Ljunggren O, Johnell O, Labrie F, function: molecular and
responsiveness of the gonadotrophs Windahl SH, Ohlsson C (2006) physiological mechanisms. Am
to sex hormone feedback in men. SHBG gene promoter J Physiol Metab 282:E601E607
J Clin Endocrinol Metab 64:6873 polymorphisms in men are Ferrini LC, Barrett-Connor E (1998)
Dobs AS, Meikle AW, Arver S, Sanders associated with serum sex hormone- Sex hormones and age: a cross-
SW, Caramelli KE, Mazer NA binding globulin, androgen and sectional study of testosterone and
(1999) Pharmacokonetics, efficacy, androgen metabolite levels, and hip estradiol and their bioavailable
and safety of a permeation- bone mineral density. J Clin fractions in community-dwelling
enhanced testosterone transdermal Endocrinol Metab 91:50295037 men. Am J Epidemiol 147:750754
system in comparison with biweekly Faber J, Perrild H, Johansen JS (1990) Field AE, Colditz GA, Willett WC,
injections of testosterone enanthate Bone Gla protein and sex hormone- Longcope C, McKinlay JB (1994)
for treatment of hypogonadal binding globulin in nontoxic goiter: The relation of smoking, age,
men. J Clin Endocrinol Metab parameters for metabolic status at relative weight and dietary intake to
84:34693478 the tissue levels. J Clin Endocrinol serum adrenal steroids, sex
Dunbar N, Gruman C, Reisine S, Metabol 70:4955 hormones and sex hormone binding
Kenny A (2001) Comparison of two Falahati-Nini A, Riggs BL, Atkinson EJ, globulin in middle age men. J Clin
health status measures and their OFallon WM, Eastell R, Khosla S Endocrinol Metab 79:13101316
associations with testosterone levels (2000) Relative contributions of Fink HA, Ewing SK, Ensrud KE,
in older men. Aging Male 4:17 testosterone and estrogen in Barrett-Connor E, Taylor BC,
Duranteau L, Chanson P, Blumberg- regulating bone resorption and Cauley JA, Orwoll ES for the
Tick J, Thomas G, Brailly S, formation in normal elderly men. Osteoporotic Fractures in Men
Lubetzki J, Schaison G, Bouchard P J Clin Invest 106:15331560 Study Group (2006) Association of
(1993) Non-responsiveness of Feldman HA, Goldstein I, testosterone and estradiol deficiency
serum gonadotropins and Hatzichristou DG, Krane RJ, with osteoporosis and rapid bone
testosterone to pulsatile GnRH in Mckinlay JB (1994) Impotence and loss in older men. J Clin Endocrinol
hemochromatosis suggesting a its medical and psychosocial Metab 91:29083915
pituitary defect. Acta Endocrinol correlates: results of the Forbes GB, Reina JC (1970) Adult lean
(Copenh) 128:351354 Massachusetts Male Aging Study. body mass declines with age: some
Elewaut A, Barbier F, Vermeulen A J Urol 151:5461 longitudinal observations.
(1979) Testosterone metabolism in Feldman HA, Longcope C, Derby CA, Metabolism 19:653663
normal males and male cirrhotics. Johannes CB, Araujo AB, Coviello Gapstur SM, Gann PH, Kopp P,
Z Gastroenterol 17:402405 AD, Bremner WJ, McKinlay JB Colangelo L, Longcope C, Liu K
Emmelot-Vonk M, Verhaar HJJ, (2002) Age trends in the level of (2002) Serum androgen
Hakhai Pour HR, Aleman A, Lock serum testosterone and other concentrations in young men: a
TMTW, Bosch JLH Ruud, Grobbee hormones in middle aged men: longitudinal analysis of
DE, van der Schouw YT (2008) longitudinal results from the associations with age, obesity and

361
Chapter 16: Androgens in male senescence

race. The CARDIA male hormone Grunstein RR, Handelsman DJ, males and females. Acta Physiol
study. Canc Epidemiol Biom Prev Lawrence SJ, Blackwell C, Caterson Scand 150:211219
11:10411047 ID, Sullivan CE (1989) Hall SA, Esche GR, Araujo AB,
Giagulli VA, Vermeulen A (1992) Neuroendocrine dysfunction in Travison TG, Clark RV,
Increased plasma 5a-androstane-3a, sleep apnea: reversal by nasal Williams RE, McKinlay JB (2008)
17b-diol glucuronide concentration continuous positive airway Correlates of low testosterone and
in clinically euthyroid women with pressure. J Clin Endocrinol Metab symptomatic androgen deficiency
suppressed plasma thyrotropin 68:352358 in a population-based sample.
levels: further evidence for Guadalupe-Grau A, Rodriguez- J Clin Endocrinol Metab 93:
generalized tissue overexposure Gonzalez FG, Ponce-Gonzalez JG, 38703877
to thyroid hormones in these Dorado C, Olmedillas H, Fuentes T,
Handelsman DJ (1994) Testicular
subjects. J Clin Endocrinol Metab Perez-Gomez J, Sanchis-moysi J,
dysfunction in systemic disease.
74:14651467 Diaz-Chico BN, Calbet JAL (2010)
Endocr Metab Clin 23:839852
Giagulli VA, Kaufman JM, Vermeulen Bone mass and the CAG and GGN
A (1994) Pathogenesis of androgen receptor polymorphisms Handelsman DJ, Dong Q (1993)
decreased androgen levels in obese in young men. Plos One 5:e11529 Hypothalamo-pituitary-gonadal
men. J Clin Endocrinol Metab Guralnik JM, Ferrucci L, Simonsick axis in chronic renal failure. Endocr
79:9971000 EM, Salive ME, Wallace RB (1995) Metab Clin 22:145161

Giusti G, Gonnelli P, Borreli D, Fiorelli Lower-extremity function in Harbitz TB (1973) Morphometric


G, Forti G, Pazzagli M, Serio M persons over 70 years as a predictor studies of Leydig cells in elderly
(1975) Age related secretion of of subsequent disability. N Engl men, with special reference to the
androstenedione, testosterone and J Med 322:556561 histology of the prostate. Acta
dihydrotestosterone by the human Haffner SM, Katz MS, Stern MP, Dunn Pathol Microbiol Scand 81:301314
testes. Exp Gerontol 10:241245 JF (1988) The relationship of sex Harkonen K, Huhtaniemi I, Makinen J,
Goldberg RB, Rabin AN, Alexander hormones to hyperinsulinemia and Hubler D, Irjala K, Koskenvuo M,
AN, Coelle GC, Getz GE (1985) hyperglycemia. Metabolism 37:683 Oettel M, Raitakari O, Saad F,
Suppression of plasma testosterone 688 Pollanen P (2003) The polymorphic
leads to an increase in serum total Haffner SM, Mykknen L, Valdez RA, androgen receptor gene CAG
and high density lipoprotein Stern MP, Katz MS (1993) repeat, pituitary-testicular function
cholesterol and apoproteins A-1 Relationship of sex hormones to and andropausal symptoms in
and B. J Clin Endocrinol Metab lipids and lipoproteins in non- ageing men. Int J Androl
60:203207 diabetic men. J Clin Endocrinol 26:187194
Gooren LJG (1987) Androgen levels Metab 77:16101615 Harman SM, Tsitouras PD (1980)
and sex functions in testosterone Haffner SM, Miettinen H, Karhapaa L, Reproductive hormones in aging
treated hypogonadal men. Arch Sex Laakso M (1997) Leptin men I. Measurment of sex steroids,
Beh 16:463476 concentrations, sex hormones and basal luteinizing hormone and
cortisol in non diabetic men. J Clin Leydig cell response to human
Gray PB, Singh AB, Woodhouse LJ,
Endocrinol Metab 82:18071809 chorionic gonadotropin. J Clin
Storer TW, Casaburi R, Dzekov J,
Endocrinol Metab 51:3540
Dzekov C, Sinha-Hikim I, Bhasin S Hajjar RR, Kaiser FE, Morley JE (1997)
(2005) Dose-dependent effects of Outcomes of long-term testosterone Harman SM, Metter EJ, Tobin JD,
testosterone on sexual function, replacement in older hypogonadal Pearson J, Blackman MR (2001)
mood, and visuospatial cognition in males: a retrospective analysis. J Clin Longitudinal effects of aging on
older men. J Clin Endocrinol Metab Endocrinol Metab 82:37933796 serum total and free testosterone
90:38383846 Hak AE, Witteman JC, de Jong FH,
levels in healthy men. J Clin
Endocrinol Metab 86:724730
Grossmann M (2011) Low testosterone Geerlings MI, Hofman A, Pols HA
in men with type 2 diabetes: (2002) Low levels of endogenous Heald AH, Ivison F, Anderson SG,
significance and treatment. J Clin androgens increase the risk of Cruickshank K, Laing I, Gibson JM
Endocrinol Metab 96: atherosclerosis in elderly men: the (2003) Significant ethnic variation
23412353 Rotterdam study. J Clin Endocrinol in total and free testosterone
Gruenewald DA, Matsumoto AM Metab 87:36323639 concentration. Clin Endocrinol
(2003) Testosterone Hkkinen K, Pakarinen A (1994) 58:262266
supplementation therapy for older Serum hormones and strength Heinemann L, Zimmermann T,
men: potential benefits and risks development during strength Vermeulen A, Thiel C, Hummel W
J Am Geriatr Soc 51:101115 training in middle-aged and elderly (1999) A new aging males

362
Chapter 16: Androgens in male senescence

symptoms rating scale. Aging Male hypothalamic-pituitary-adrenal regulation of estrogen biosynthesis


2:105114 functioning in normal males. Drug in humans. Trends Endocrinol
Hollander N, Hollander VP (1958) The Alcohol Dep 31:5764 Metab 13:122128
microdetermination of testosterone Impallomeni M, Kaufman BM, Palmer Kamischke A, Kemper DE, Castel MA,
in human spermatic vein blood. AJ (1994) Do acute diseases Luthke M, Rolf C, Behre HM,
J Clin Endocrinol Metab 18:966970 transiently impair anterior pituitary Magnussen H, Nieschlag E (1998)
Howell SJ, Radford JA, Adams JE, function in patients over the age of Testosterone levels in men with
Smets EM, Warburton R, Shalet SM 75? A longitudinal study of the TRH chronic obstructive pulmonary
(2001) Randomized placebo- test and basal gonadotrophin levels. disease with or without
controlled trial of testosterone Postgrad Med J 70:8691 glucocorticoid therapy. Eur Respir J
replacement in men with mild Irwin M, Dreyfus E, Baird DS, Smith T, 11:4145
Leydig cell insufficiency following Schuckit M (1988) Testosterone in Katznelson L, Finkelstein JS,
cytotoxic chemotherapy. Clin chronic alcoholic men. Br J Addict Schoenfeld DA, Rosenthal DI,
Endocrinol (Oxf) 55:315324 83:949953 Anderson EJ, Klibanski A (1996)
Huhtaniemi IT, Pye SR, Limer KL, Isidori AM, Giannetta E, Gianfrilli D, Increase in bone density and lean
Thomson W, ONeill TW, Platt H, Greco EA, Bonifacio V, Aversa A, body mass during testosterone
Payne D, John SL, Jiang M, Boonen Isidori A, Fabbri A, Lenzi A (2005a) administration in men with
S, Borghs H, Vanderschueren D, Effects of testosterone on sexual acquired hypogonadism. J Clin
Adams JE, Ward KA, Bartfai G, function in men: results of a meta- Endocrinol Metab 81:43584365
Casanueva F, Finn JD, Forti G, analysis. Clin Endocrinol Kaufman JM, Goemaere S (2008)
Giwercman A, Han TS, Kula K, 63:381394 Osteoporosis in men. Best Pract Res
Lean MEJ, Pendleton N, Punab M, Isidori AM, Giannetta E, Greco EA, Clin Endocrinol Metab 22:787812
Silman AJ, Wu FCW; EMAS Study Gianfrilli D, Bonifacio V, Isidori A, Kaufman JM, Vermeulen A (1997)
Group (2009) Increased estrogen Lenzi A, Fabbri A (2005b) Effects of Declining gonadal function in
rather than decreased androgen testosterone on body composition, elderly men. Baillires Endocrinol
action is associated with longer bone metabolism and serum lipid Metab 11:289309
androgen receptor CAG repeats. profile in middle-aged men: a meta-
J Clin Endocrinol Metab 94:277284 analysis. Clin Endocrinol
Kaufman JM, Vermeulen A (2005)
The decline of androgen levels in
Huhtaniemi IT, Pye SR, Holliday KL, 63:280293
elderly men and its clinical and
Thomson W, ONeill TW, Platt H, Jackson JA, Waxman J, Spiekerman M therapeutic implications. Endocrine
Payne D, John SL, Jiang M, Bartfai (1989) Prostatic implications of Rev 26:833876
G, Boonen S, Casanueva FF, Finn testosterone replacement therapy.
JD, Forti G, Giwercman A, Han TS, Kaufman JM, Giri M, Deslypere JP,
Arch Intern Med 149:23642366
Kula K, Lean MEJ, Pendleton N, Thomas G, Vermeulen A (1991)
Punab M, Silman AJ, Jackson JA, Riggs MW, Spiekerman M Influence of age on the
Vanderschueren D, Labrie F, Wu (1992) Testosterone deficiency as a responsiveness of the gonadotrophs
FCW; European Male Aging Study risk factor for hip fracture in men: a to luteinizing hormone-releasing
Group (2010) Effect of case control study. Am J Med Sci hormone in males. J Clin Endocrinol
polymorphisms in selected genes 304:48 Metab 72:12551260
involved in pituitary-testicular Joshi D, van Schoor NM, de Ronde W, Kenny A, Kleppinger A, Annis K,
function on reproductive hormones Schaap LA, Comijs HC, Beekman Rathier M, Browner B, Judge JO,
and phenotype in aging men. J Clin ATF, Lips P (2010) Low free McGee D (2010) Effects of
Endocrinol Metab 95:18981908 testosterone levels are associated transdermal testosterone on bone
Hyde Z, Flicker L, Almeida OP, with prevalence and incidence of and muscle in older men with low
Hankey GJ, McCaul KA, Chubb depressive symptoms in older men. bioavailable testosterone levels, low
SAP, Yeap BB (2010) Low free Clin Endocrinol 72:232240 bone mass, and physical frailty.
testosterone predicts frailty in older Jung A, Schuppe HC, Schill WB (2002) JAGS 58:11341143
men: the health in men study. J Clin Comparison of semen quality in Kenny AM, Prestwood KM, Gruman
Endocrinol Metab 95:31653172 older and younger men attending an CA, Marcello KM, Raisz LG (2001)
Ida Y, Tsyjimaru S, Nakamura K, andrology clinic. Andrologia Effects of transdermal testosterone
Shirao I, Mukasa H, Egarni H, 34:116122 on bone and muscle in older men
Nakazawa H (1992) Effect of acute Kamat A, Hinshelwood MM, Murry with low bioavailable testosterone
and repeated alcohol ingestion on BA, Mendelson CR (2002) levels. J Gerontol A Biol Sci Med Sci
hypothalamic-pituitary-gonadal and Mechanisms in tissue-specific 56:M266M272

363
Chapter 16: Androgens in male senescence

Kent JZ, Acone AB (1966) Plasma Kupelian V, Hayes FJ, Link CL, Rosen Lindstrm S, Ma J, Altshuler D,
androgens and aging. In: R, McKinlay JB (2008) Inverse Giovannucci E, Riboli E, Albanes D,
Vermeulen A, Exley D (eds) association of testosterone and the Allen NE, Berndt SI, Boeing H,
Androgens in Normal and metabolic syndrome in men is Bueno-de-Mesquita HB, Chanock
Pathological Conditions. Excerpta consistent across race and ethnic SJ, Dunning AM, Feigelson HS,
Medica Foundation, Amsterdam, groups. J Clin Endocrinol Metab Gaziano JM, Haiman CA, Hayes
pp 3135 93:34033410 RB, Henderson BE, Hunter DJ,
Khaw KT, Barrett-Connor E (1992) Kwan M, Greenleaf WJ, Mann J, Crapo Kaaks R, Kolonel LN, Le Marchand
Lower endogenous androgens L, Davidson J (1983) The nature of L, Martinez C, Overvad K, Siddiq A,
predict central obesity in men. Ann androgen action on male sexuality: Stampfer M, Stattin P, Stram DO,
Epidemiol 2:675682 a combined laboratory/self Thun MJ, Trichopoulos D,
report study on hypogonadal men. Tumino R, Virtamo J, Weinstein SJ,
Khosla S (2010) Update in male Yeager M, Kraft P, Freedman ML
osteoporosis. J Clin Endocrinol J Clin Endocrinol Metab
57:557562 (2010) A large study of androgen
Metab 95:310 receptor germline variants and their
Khosla S, Melton LJ III, Atkinson EJ, Labrie F, Blanger A, Simard J, Van relation to sex hormone levels and
OFallon WM (2001) Relationship Luu-The, Labrie C (1995) DHEA prostate cancer risk. Results from
of serum sex steroid levels to and peripheral androgen and the National Cancer Institute Breast
longitudinal changes in bone estrogen formation: intracrinology. and Prostate Cancer Cohort
density in young versus elderly men. Ann N Y Acad Sci 774:1628 Consortium. J Clin Endocrinol
J Clin Endocrinol Metab 86: Lakshman KM, Kaplan B, Travison Metab 95:E121E127
35553561 TG, Basaria S, Knapp PE, Singh AB, Liu PY, Death AK, Handelsman DJ
Korenman SG, Morley JE, Mooradian La Valley MP, Mazer NA, Bhasin S (2003) Androgens and
AD, Davis SS, Kaiser FE, Silver AJ, (2010) The effects of injected cardiovascular disease. Endocr Rev
Viosca SP, Garza D (1990) testosterone dose and age on the 24:313340
Secondary hypogonadism in older conversion of testosterone to
estradiol and dihydrotestosterone in Liu PY, Beilin J, Meier C, Nguyen TV,
men: its relation to impotence. Center JR, Leedman PJ, Seibel MJ,
J Clin Endocrinol Metab 71:963969 young and older men. J Clin
Endocrinol Metab 95: Eisman JA, Handelsman DJ (2007a)
Krasnoff JB, Basaria S, Pencina MJ, 39553964 Age-related changes in serum
Jasuja GK, Vasan RS, Ulloor J, testosterone and sex hormone
Zhang A, Coviello A, Kelly-Hayes Lange JD, Brown WA, Wincze J, Zwick binding globulin in Australian men:
M, DAgostino RB, Wolf PA, Bhasin W (1980) Serum testosterone longitudinal analyses of two
S, Murabito JM (2010) Free concentration and penile geographically separate regional
testosterone levels are associated tumescence changes in men. Horm cohorts. J Clin Endocrinol Metab
with mobility limitation and Behav 14:267270 92:35993603
physical performance in Lapauw B, Goemaere S, Zmierczak H, Liu PY, Caterson ID, Grunstein RR,
community-dwelling men: the Van Pottelbergh I, Mahmoud A, Handelsman DJ (2007b)
Framingham Offspring Study. J Clin Taes Y, De Bacquer D, Vansteelandt Androgens, obesity, and sleep-
Endocrinol Metab 95:27902799 S, Kaufman JM (2008) The decline disordered breathing in men.
Kunelius P, Lukkarinen O, Hannuksela of serum testosterone levels in Endocrinol Metab Clin N Am
ML, Itkonen O, Tapanainen JS community-dwelling men over 70 36:349363
(2002) The effects of transdermal years of age: descriptive data and
predictors of longitudinal changes. Longcope C (1973) The effect of
dihydrotestosterone in the aging human chorionic gonadotropin
male: a prospective, randomized, Eur J Endocrinol 159:459468
on plasma steroid levels in
double blind study. J Clin Lazarou S, Morgentaler A (2008) The young and old men. Steroids
Endocrinol Metab 87:14671472 effect of aging on spermatogenesis 21:583592
Kupelian V, Page ST, Araujo AB, and pregnancy outcomes. Urol Clin
Travison TG, Bremner WJ, N Am 35:331339 Longcope C, Feldman HA, McKinlay
JB, Araujo AB (2000) Diet and sex
McKinlay JB (2006) Low sex Lichtenstein MJ, Yarnell JWG, Elwood
hormone-binding globulin. J Clin
hormone-binding globulin, total PC, Beswick AD, Sweetnam PM,
Endocrinol Metab 85:293296
testosterone, and symptomatic Marks V, Teale D, Riad-Fahmy D
androgen deficiency are associated (1987) Sex hormones, insulin, lipid Luboshitzky R, Aviv A, Hefetz A, Herer
with development of the metabolic and prevalent ischemic heart P, Shen-Orr Z, Lavie L, Lavie P
syndrome in nonobese men. J Clin disease. Am J Epidemiol (2002) Decreased pituitary-gonadal
Endocrinol Metab 91:843850 126:647657 secretion in men with obstructive

364
Chapter 16: Androgens in male senescence

sleep apnea. J Clin Endocrinol Metab genetic and non genetic factors to on testosterone levels in older men:
87:33943398 determine plasma sex steroid longitudinal results from the
variation in normal male twins. Massachusetts male ageing study.
Ly LP, Jimenez M, Zhuang TN,
Metabolism 35:10901095 Eur J Endocrinol 155:443452
Celermajer DS, Conway AJ,
Handelsman DJ (2001) A double- Meikle AW, Stringham JD, Bishop T, Mohr BA, Bhasin S, Kupelian V,
blind, placebo-controlled, West DW (1988) Quantitation of Araujo AB, ODonnell AB,
randomized clinical trial of genetic and nongenetic factors McKinlay JB (2007) Testosterone,
transdermal dihydrotestosterone gel influencing androgen productions sex hormone-binding globulin, and
on muscular strength, mobility, and and clearance rates in men? J Clin frailty in older men. J Am Geriatr
quality of life in older men with Endocrinol Metab 67: Soc 55:548555
partial androgen deficiency. J Clin 104109 Moorjani S, Dupont A, Labrie F,
Endocrinol Metab 86:40784088 Mellstrm D, Vandenput L, Mallmin Lupien PJ, Brun D, Gagn C,
MacAdams MR, White RH, Chipps BE H, Holmberg A, Lorentzon M, Gigure M, Blanger A (1987)
(1986) Reduction of serum Oden A, Johansson H, Orwoll ES, Increase in plasma high density
testosterone levels during chronic Labrie F, Karlsson MK, Ljunggren lipoprotein concentration following
glucocorticoid therapy. Ann Intern O, Ohlsson C (2008) Older men complete androgen blockade in men
Med 104:648651 with low serum estradiol and high with prostatic carcinoma.
serum SHBG have an increased risk Metabolism 36:244250
Mahmoud AM, Goemaere S, De of fractures. J Bone Miner Res
Bacquer D, Comhaire FH, Kaufman Morley JE (1986) Impotence. Am J Med
23:15521560 80:897906
JM (2000) Serum inhibin B levels in
community-dwelling elderly men. Mikuma N, Kumamoto Y, Maruta H, Morley JE, Kaiser FE, Perry HM III,
Clin Endocrinol 53:141147 Nitta T (1994) Role of the Patrick P, Morley PM, Stauber PM,
hypothalamic opioidergic system in Vellas B, Baumgartner RN, Garry PJ
Mahmoud AM, Goemaere S, El-Garem the control of gonadotropin
Y, Van Pottelbergh I, Comhaire FH, (1997) Longitudinal changes in
secretion in elderly men. Andrologia testosterone, luteinizing hormone
Kaufman JM (2003) Testicular 26:3945
volume in relation to hormonal and follicle stimulating hormone in
indices of gonadal function in Mitchell R, Hollis S, Rothwell C, healthy older men. Metabolism
community-dwelling elderly men. Robertsons WR (1995) Age related 46:410413
J Clin Endocrinol Metab 88:179184 changes in the pituitary testicular Morley JE, Charlton E, Patrick P,
axis in normal men; lower serum Kaiser FE, Cadeau P, McCready D,
Maki PM, Ernst M, London ED, testosterone results from decreased
Mordecai KL, Perschler P, Durso Perry HM III (2000) Validation of a
bioactive LH drive. Clin Endocrinol screening questionnaire for
SC, Brandt J, Dobs A, Resnick SM 42:501507
(2007) Intramuscular testosterone androgen deficiency in aging males.
treatment in elderly men: evidence Mittan D, LEE S, Miller E, Perez RC, Metabolism 49:12391242
of memory decline and altered brain Basler JW, Bruder JM (2002) Bone Morley JE, Patrick P, Perry HM III
function. J Clin Endocrinol Metab loss following hypogonadism in (2002) Evaluation of assays available
92:41074114 men with prostate cancer treated to measure free testosterone.
with GnRH analogs. J Clin Metabolism 51:554559
Masters WH (1986) Sex and aging Endocrinol Metab 87:36563661
expectations and reality. Hosp Pract Muller M, Grobbee DE, Thijssen JHH,
Moffat SD, Zonderman AB, Metter EJ,
15:175198 van den Beld AW, van der Schouw
Kawas C, Blackman MR, Harman YT (2003) Sex hormones and male
Matzkin H, Braf Z, Nava D (1991) SM, Resnick SM (2004) Free health: effects on components of the
Does age influence the bioactivity of testosterone and risk for Alzheimer frailty syndrome. Trends Endocrinol
follicle-stimulating hormone in disease in older men. Neurology Metab 14:289296
men. Age Ageing 20:199205 62:188193
Mulligan T, Iranmanesh A, Gheorghiu
Meier C, Nguyen TV, Handelsman DJ, Mohr BA, Guay AT, ODonnell AB, S, Godschalk M, Veldhuis JD (1995)
Schindler C, Kushnir MM, McKinlay JB (2005) Normal, bound Amplified nocturnal luteinizing
Rockwood AL, Meikle AW, Center and nonbound testosterone levels in hormone (LH) secretory burst
JR, Eisman JA, Seibel MJ (2008) normally ageing men: results from frequency with selective attenuation
Endogenous sex hormones and the Massachusetts Male Ageing of pulsatile (but not basal)
incident fracture risk in older men. Study. Clin Endocrinol 62:6473 testosterone secretion in healthy
Arch Intern Med 168:4754 Mohr BA, Bhasin S, Link CL, aging men: possible Leydig cell
Meikle AW, Bishop DT, Stringham JD, ODonnell AB, Mckinlay JB (2006) desensitization to endogenous LH
West DW (1986) Quantitating The effect of changes in adiposity signaling-a clinical research center

365
Chapter 16: Androgens in male senescence

study. J Clin Endocrinol Metab OConnell MDL, Roberts SA, Srinivas- meta-analysis. J Am Geriatr Soc
80:30253031 Shankar U, Tajar A, Connolly MJ, 54:16661673
Mulligan T, Iranmanesh A, Kerzner R, Adams JE, Oldham JA, Wu FCW Perry PJ, Lund BC, Arndt S, Holman T,
Demers LW, Veldhuis JD (1999) (2011) Do the effects of testosterone Bever-Stille KA, Paulsen J, Demers
Two-week pulsatile gonadotropin on muscle strength, physical LM (2001) Bioavailable testosterone
releasing hormone infusion function, body composition, and as a correlate of cognition,
unmasks dual (hypothalamic and quality of life persist six months psychological status, quality of life,
Leydig cell) defects in the healthy after treatment in intermediate-frail and sexual function in aging males:
aging male gonadotropic axis. and frail elderly men? J Clin implications for testosterone
Europ J Endocrinol 141:257266 Endocrinol Metab 96:454458 replacement therapy. Ann Clin
Mulligan T, Iranmanesh A, Veldhuis Orwoll E, Lambert LC, Marshall LM, Psychiatry 13:7580
JD (2001) Pulsatile iv infusion of Phipps K, Blank J, Barrett-Connor Pfeilschifter J, Scheidt-Nave C, Leidig-
recombinant human LH in E, Cauley J, Ensrud K, Cummings S, Bruckner G, Woitge HW, Blum
leuprolide-suppressed men for the Osteoporotic Fractures in WF, Wster C, Haack D, Ziegler R
unmasks impoverished Leydig-cell Men Study Group (2006a) (1996) Relationship between
secretory responsiveness to Testosterone and estradiol among circulating insulin-like growth
midphysiological LH drive in the older men. J Clin Endocrinol Metab factor components and sex
aging male. J Clin Endocrinol Metab 91:13361344 hormones in a population based
86:55475553 Orwoll E, Lambert LC, Marshall LM, sample of 5080 year old men and
Blank J, Barrett-Connor E, Cauley J, women. J Clin Endocrinol Metab
Mnzer T, Haeman MS, Hees P,
Shapiro E, Christmas C, Bellantoni
Ensrud K, Cummings SR, for the 81:25342540
Osteoporotic Fractures in Men
MF, Stevens TE, OConnor KG, Phillips GB, Pinkernell BJ, Jing TY
Study Group (2006b) Endogenous
Pabst KM, Clair C Sr, Sorkin JD, (1994) The association of
testosterone levels, physical
Blackman MR (2001) Effects of GH hypotestosteronemia with coronary
performance, and fall risk in
and/or sex steroid administration heart disease. Arterioscler Thromb
older men. Arch Intern Med
on abdominal subcutaneous and 14:701706
166:21242131
visceral fat in healthy aged women Pincus SM, Veldhuis JD, Mulligan T,
and men. J Clin Endocrinol Metab Orwoll ES, Klein RF (1995)
Iranmanesh A, Evans WS (1997)
86:36043610 Osteoporosis in men. Endocr Rev
Effects of age on the irregularity of
16:87115
Nankin HR, Lin T, Murono EP, LH and FSH serum concentrations
Osterman J (1981) The aging Leydig Orwoll ES, Oviatt S (1992) in women and men. Am J Physiol
cell III. Gonadotropin stimulation Transdermal testosterone 273:E989E995
in men. J Androl 2:181189 supplementation in normal older
Pirke KM, Sintermann R, Vogt HJ
men. In: Program of the 74th
Neaves WB, Johnson L, Porter JC, (1980) Testosterone and
Annual Meeting of the Endocrine
Parker CR, Petty CS (1984) Leydig testosterone precursors in the
Society. The Endocrine Society,
cell numbers, daily sperm spermatic vein and in the testicular
Bethesda; Abstract 1071
production and gonadotropin levels tissue of old men. Gerontology
in aging men. J Clin Endocrinol Orwoll ES, Nielson CM, Labrie F, 26:221230
Metab 59:756763 Barrett-Connor E, Cauley JA,
Cummings SR, Ensrud K, Karlsson Plymate SR, Tenover JS, Bremner WJ
Ng KK, Donat R, Chan L, Lalak A, Di M, Lau E, Leung PC, Lunggren O, (1989) Circadian variation in
Pierro I, Handelsman DJ (2004) Mellstrm D, Patrick A, Stefanick testosterone, sex hormone-binding
Sperm output of older men. Hum ML, Nakamura K, Yoshimura N, globulin, and calculated non sex
Reprod 19:18111815 Zmuda J, Vandenput L, Ohlsson C hormone-binding globulin bound
Nieschlag E, Kley KH, Wiegelmann W, and for the Osteoporotic testosterone in healthy young and
Solback HG, Krskemper HL Fractures in Men (MrOS) Research elderly men. J Androl 10:366371
(1973) Lebensalter und endokrine Group (2010) Evidence for Poggi UI, Arguelles AE, Rosner J, de
Funktion der Testes des geographical and racial variation in Laborde NP, Cassini JH, Volmer
erwachsenen Mannes. Deut Med serum sex steroid levels in older MC (1976) Plasma testosterone and
Wschr 98:12811284 men. J Clin Endocrinol Metab 95: serum lipids in male survivors of
Nieschlag E, Lammers U, Freischem E151E160 myocardial infarction. J Steroid
CW, Langer K, Wickings EJ (1982) Ottenbacher KJ, Ottenbacher ME, Biochem 7:229231
Reproductive functions in young Ottenbacher AJ, Acha AA, Ostir GV Porto CS, Abreu LC, Gunsalus GL,
fathers and grandfathers. J Clin (2006) Androgen treatment and Bardin CW (1992) Binding of sex
Endocrinol Metab 55:676681 muscle strength in elderly men: a hormone binding globulin (SHBG)

366
Chapter 16: Androgens in male senescence

to testicular membranes and Rohrmann S, Platz EA, Selvin E, Shielst Ruhayel Y, Malm G, Haugen TB,
solubilized receptor. Mol Cell MS, Joshut CE, Menket A, Feinleibt Henrichsen T, Bjsvikt C,
Endocrinol 89:3338 M, Basaria S, Rifai N, Dobs AS, Grotmol T, Saethert T, Malm J,
Rajagopal A, Vassilopoulou-Sellin R, Kanarek N, Nelson WG (2011) The Figenschau Y, Rylander L, Levine
Palmer JL, Kaur G, Bruera E (2004) prevalence of low sex steroid RJ, Giwercman A (2007) Seasonal
Symptomatic hypogonadism in hormone concentrations in men in variation in serum concentrations
male survivors of cancer with the Third National Health and of reproductive hormones and
chronic exposure to opioids. Cancer Nutrition Examination Survey urinary excretion of 6-
100:851858 (NHANES III). Clin Endocrinol sulfatoxymelatonin in men living
75:232239 north and south of the Arctic Circle:
Rajfer JK, Namkun PC, Petra Ph (1989) a longitudinal study. Clin
Identification, partial Rolf C, Behre M, Nieschlag E (1996)
Reproductive parameters of older Endocrinol 67:8592
characterization and age associated
changes of a cytoplasmatic compared to younger men of Ruige JB, Mahmoud AM, De Bacquer D,
androgen receptor in the rat infertile couples. Int J Androl Kaufman JM (2011) Endogenous
penis. J Steroid Biochem 33: 19:135142 testosterone and cardiovascular
14891492 Rolland Y, Dupuy C, Abellan van Kan disease in healthy men: a meta-
G, Gillette S, Vellas B (2011) analysis. Heart 97:870875
Rhoden EL, Teloken C, Mafessoni R,
Souto CA (2002) Is there any Treatment strategies for sarcopenia Russell DW, Wilson JD (1994) Steroid
relation between serum levels of and frailty. Med Clin N Am 5a-reductase: two genes/two
total testosterone and the severity of 95:427438 enzymes. Annu Rev Biochem
erectile dysfunction? Int J Impot Res Rosner W, Hryb DJ, Kahn MS, Nakhla 63:2561
14:167171 HM, Romas NA (1991) Sex Sandblom RE, Matsumoto AM, Scoene
Ring HZ, Lessov CN, Reed T, hormone binding globulin: RB, Lee KA, Giblin EC, Bremner
Marcux R, Holloway L, Swan GE, anatomy and physiology of a new WJ, Pierson DJ (1983) Obstructive
Carmelli D (2005) Heritability of regulating system. J Steroid Biochem sleep apnea induced by testosterone
plasma sex hormones and hormone Mol Biol 40:813820 administration N Engl J Med
binding globulin in adult male Rosner W, Auchus RJ, Azziz R, Sluss 308:508510
twins. J Clin Endocrinol Metab PM, Raff H (2007) Position Sartorius G, Spasevska S, Idan A,
90:36533658 statement: utility, limitations, and Turner L, Allan L, Ly LP, Conway
Rodriguez A, Muller DC, Metter EJ, pitfalls in measuring testosterone: AJ, McLachlan RI, Handelsman DJ
Maggio M, Harman SM, Blackman an Endocrine Society position (2011) Older men reporting
MR, Andres R(2007) Aging, statement. J Clin Endocrinol Metab excellent asymptomatic health
androgens, and the metabolic 92:405413 exhibit no decline in serum
syndrome in a longitudinal study of Roy TA, Blackman MR, Harman SM, testosterone, dihydrotestosterone or
aging. J Clin Endocrinol Metab Tobin JD, Schrager M, Metter EJ estradiol with age: the healthy man
92:35683572 (2002) Interrelationships of serum study. Endocr Rev 32:OR351
testosterone and free testosterone (abstract)
Roehrborn CG, Lange JL, George FW,
Wilson JD (1987) Changes in index with FFM and strength in Sasano N, Ichijo S (1969) Vascular
amount and intracellular aging men. Am J Physiol Endocrinol patterns of the human testes with
distribution of androgen receptor in Metab 283:E284E294 special reference to its senile
human foreskin as a function of age. Rubens R, Dhont M, Vermeulen A changes. Tohoku J Exp Med
J Clin Invest 79:4447 (1974) Further studies on Leydig 99:269280
Rogers MA, Evans WJ (1993) Changes cell function in old age. J Clin Sattler FR, Castaneda-Sceppa C, Binder
in skeletal muscle with aging: effects Endocrinol Metab 39:4045 EF, Schroeder ET, Wang Y, Bhasin
of exercise training. Exercise Sports Rubenstein LZ, Josephson KR, Robbins S, Kawakubo M, Stewart Y,
Sci Rev 21:65102 AS (1994) Falls in the nursing Yarasheski KE, Ulloor J, Colletti P,
Rohrmann S, Nelson WG, Rifai N, home. Ann Intern Med 121: Roubenoff R, Azen SP (2009)
Brown TR, Dobs A, Kanarek N, 442451 Testosterone and growth hormone
Yager JD, Platz EA (2007) Serum Rubin HB, Henson DE, Falvo RE, High improve body composition and
estrogen, but not testosterone, levels RW (1979) The relationship muscle performance in older men.
differ between black and white men between mens endogenous levels of J Clin Endocrinol Metab
in a nationally representative testosterone and their penile 94:19912001
sample of Americans. J Clin response to erotic stimuli. Behav Res Schaap LA, Pluijm SMF, Deeg DJH,
Endocrinol Metab 92:25192525 Ther 17:305312 Penninx BW, Nicklas BJ, Lips P,

367
Chapter 16: Androgens in male senescence

Harris TB, Newman AB, in plasma testosterone levels in related to disease severity. J Clin
Kritchevsky SB, Cauley JA, man. J Clin Endocrinol Metab Endocrinol Metab 76:15481554
Goodpaster BH, Tylavsky FA, 42:979982 Srinivas-Shankar U, Roberts SA,
Yaffe K, Visser M (2008) Low Smith KW, Gruman C, Reisine S, Connolly MJ, OConnell MDL,
testosterone levels and decline in Kenny A (2000) Construction and Adams JE, Oldham JA, Wu FCW
physical performance and muscle field validation of a self- (2010) Effects of testosterone on
strength in older men: findings administered screener for muscle strength, physical function,
from two prospective cohort testosterone deficiency body composition, and quality of
studies. Clin Endocrinol 68:4250 (hypogonadism) in aging men. Clin life in intermediate-frail and frail
Schiavi RC, Schreiner-Engel P, Endocrinol (Oxf) 53:703711 elderly men: a randomized, double-
Mandeli J, Schanzer H, Cohen E Sniffen RC (1950) The testes. blind, placebo-controlled study.
(1990) Healthy aging and male I. The normal testis. Arch Pathol J Clin Endocrinol Metab 95:639650
sexual function. Am J Psychiatry 50:259284 Stanley HL, Schmitt BP, Poses RM,
147:776771 Deiss WP (1991) Does
Snyder PJ, Peachey H, Berlin JA,
Schneider HJ, Sievers C, Klotsche J, Hannoush P, Berlin JA, Loh L, hypogonadism contribute to the
Bhler S, Pittrow D, Lehnert H, Lendrow DA, Holmes JH, Dlewati occurrence of a minimal trauma hip
Wittchen HU, Stalla GK (2009) A, Santanna J, Rosen CJ, Strom BL fracture in elderly men? JAGS
Prevalence of low male testosterone (1999a) Effect of testosterone 39:766771
levels in primary care in Germany: treatment on body composition and Stanworth RD, Kapoor D, Channer KS,
cross-sectional results from the muscle strength in men over 65 Jones TH (2008) Androgen receptor
DETECT study. Clin Endocrinol years of age. J Clin Endocrinol CAG repeat polymorphism is
70:446454 Metab 84:26472653 associated with serum testosterone
Schrder FH (1996) The prostate and Snyder PJ, Peachey H, Berlin JA, levels, obesity and serum leptin in
androgens: the risk of Hannoush P, Berlin JA, Loh L, men with type 2 diabetes. Eur
supplementation. In: Oddens B, Holmes JH, Dlewati A, Staley J, J Endocrinol 159:739746
Vermeulen A (eds) Androgens and Santanna J, Kapoor SC, Attie MF, Steidle C, Schwartz S, Jacoby K, Sebree
the Aging Male. Parthenon Haddad JG Jr, Strom BL (1999b) T, Smith T, Bachand R (2003)
Publishing Group, New York, pp Effect of testosterone treatment on AA2500 testosterone gel normalizes
223226 bone mineral density in men over androgen levels in aging males with
Semple PD, Beastall GH, Brown TM, 65 years of age. J Clin Endocrinol improvements in body composition
Stirling KW, Mills RJ, Watson WS Metab 84:19661972 and sexual function. J Clin
(1984) Sex-hormone suppression and Snyder PJ, Peachey H, Berlin JA, Endocrinol Metab 88:26732681
sexual impotence in hypoxic Hannoush P, Haddad G, Dlewati A, Stoch AS, Parker RA, Chen L, Bubley
pulmonary fibrosis. Thorax 39:4651 Santanna J, Loh L, Lenrow DA, G, Ko Y-J, Vincelette A, Greenspan
Shahinian VB, Kuo Y, Freeman JL, Holmes JH, Kapoor SC, Atkinson SL (2001) Bone loss in men with
Goodwin JS (2005) Risk of fracture LE, Strom BL (2000) Effects of prostate cancer treated with
after androgen deprivation for testosterone replacement in gonadotropin-releasing hormone
prostate cancer. New Engl J Med hypogonadal men. J Clin Endocrinol agonists. J Clin Endocrinol Metab
352:154164 Metab 85:26702677 86:27872791
Sih R, Morley JE, Kaiser FE, Perry Sparrow D, Silbert JE, Rowe JW (1980) Stripp B, Taylor AA, Bartter FC,
HM III, Patrick P, Ross C (1997) The influence of age, alcohol Gillette JR, Loriaux DL, Easley R,
Testosterone replacement in older consumption, and body build on Menard RH (1975) Effect of
hypogonadal men: a 12 month gonadal function in men. J Clin spironolactone on sex hormones in
randomized controlled trial. J Clin Endocrinol Metab 51:508512 man. J Clin Endocrinol Metab
Endocrinol Metab 82:16611667 Spratt DI, ODea L, Schoenfeld D, 41:777781
Siiteri PK, MacDonald PC (1975) Role Butler J, Narashimha H, Rao P, Suoranta H (1971) Changes in small
of extraglandular estrogen in Crowley WF (1988) vessels of the adult testes in relation
human endocrinology. In: Greep Neuroendocrine-gonadal axis in to age and some pathological
RD, Astwood B (eds) Handbook of men: frequent sampling of LH, FSH conditions. Virchows Arch A Pathol
Physiology, Vol. II. American and testosterone. Am J Physiol 254: Pathol Anat 352:765781
Physiological Society, Washington, E658E666 Svartberg J, Jorde R, Sundsfjord J,
DC, pp 491508 Spratt DI, Cox P, Orav J, Moloney J, Bnaa KH, Barrett-Connor E (2003)
Smals AGH, Kloppenburg PWC, Bigos T (1993) Reproductive axis Seasonal variation of testosterone
Benraad TJ (1976) Circannual cycle suppression in acute illness is and waist to hip ratio in men: the

368
Chapter 16: Androgens in male senescence

Troms Study. J Clin Endocrinol Tracz MJ, Sideras K, Bolona ER, aromatase inhibition. J Clin
Metab 88:30993104 Haddad RM, Kennedy CC, Uraga Endocrinol Metab 90:57175722
Swartz CM, Young MA (1987) Low MV, Caples SM, Erwin PJ, Montori TSjoen GG, De Vos S, Goemaere S,
serum testosterone and myocardial VM (2006) Testosterone use in men Van Pottelbergh I, Dierick M, Van
infarction in geriatric male patients. and its effects on bone health. Heeringen C, Kaufman JM (2005b)
J Am Geriatr Soc 35:3944 A systematic review and meta- Sex steroid level, androgen receptor
analysis of randomized placebo- polymorphism, and depressive
Tajar A, Forti G, ONeill TW, Lee DM, controlled trials. J Clin Endocrinol
Silman AJ, Finn JD, Bartfai G, symptoms in healthy elderly men.
Metab 91:20112016 J Am Geriatr Soc 53:636642
Boonen S, Casanueva FF,
Giwercman A, Han TS, Kula K, Travison TG, Morley JE, Araujo AB, Turner HE, Wass JAH (1997) Gonadal
Labrie F, Lean MEJ, Pendleton N, ODonnell AB, McKinlay JB (2006) function in men with chronic
Punab M, Vanderschueren D, The relationship between libido illness. Clin Endocrinol 47:379403
Huhtaniemi IT, Wu FCW; EMAS and testosterone levels in aging
men. J Clin Endocrinol Metab Udry JR, Billy JO, Morris NM, Groff
Group (2010) Characteristics of TR, Raj MH (1985) Serum
secondary, primary, and 91:25092513
androgenic hormones motivate
compensated hypogonadism in Travison TG, Araujo AB, Kupelian V, normal behavior in adolescent boys.
aging men: evidence from the ODonnell AB, McKinlay JB (2007) Fertil Steril 43:9094
European Male Ageing Study. J Clin The relative contributions of
Urban RJ, Veldhuis JD, Blizzard R,
Endocrinol Metab 95:18101818 aging, health, and lifestyle factors
Dufau ML (1988) Attenuated
Tapia-Serrano R, Jimenez-Baldera FJ, to serum testosterone decline in
release of biologically active
Murrieta S, Bravo-Gatica C, Guerra men. J Clin Endocrinol Metab
luteinizing hormone in healthy
R, Mintz G (1991) Testicular 92:549555
aging men. J Clin Invest 81:
function in active ankylosing Travison TG, Nguyen AH, Naganathan 10201029
spondylitis therapeutic response V, Stanaway FF, Blyth FM,
Vanbillemont G, Bogaert V, De
to human chorionic gonadotrophin. Cumming RG, Le Couteur DG,
Bacquer D, Lapauw B, Goemaere S,
J Rheumatol 18:841848 Sambrook PN, Handelsman DJ
Toye K, Van Steen K, Taes Y,
Tenover JS (1994) Androgen (2011) Changes in reproductive
Kaufman JM (2009)
administration to aging men. hormone concentrations predict the
Polymorphisms of the SHBG gene
Endocrinol Metab Clin N Am prevalence and progression of the
contribute to the interindividual
23:877889 frailty syndrome in older men: the
variation of sex steroid hormone
concord health and ageing in men
Tenover JS, Matsumoto AM, Plymate blood levels in young, middle-aged
project. J Clin Endocrinol Metab
SR, Bremner WJ (1987) The effects and elderly men. Clin Endocrinol
96:24642474
of aging in normal men on 70:303310
bioavailable testosterone and Tsitouras PD, Bulat T (1995) The aging
van den Beld AW, de Jong FH,
luteinizing hormone secretion: male reproductive system.
Grobbee DE, Pols HAP, Laberts
response to clomiphene Endocrinol Metab Clin 24:297315
SWJ (2000) Measures of
citrate. J Clin Endocrinol Metab Tsitouras PD, Martin CE, Harman SM bioavailable serum testosterone and
65:11181126 (1982) Relation of serum estradiol and their relationships
Tenover JS, Matsumoto AM, Clifton testosterone to sexual activity in with muscle strength, bone density,
DK, Bremmer WJ (1988) Age healthy elderly men. J Gerontol and body composition in elderly
related alterations in the circadian 37:288293 men. J Clin Endocrinol Metab
rhythms of pulsatile luteinizing TSjoen G, Goemaere S, De Meyere M, 85:32763282
hormone and testosterone secretion Kaufman JM (2004) Perception of van den Beld AW, Bots ML, Janssen JA,
in healthy men. J Gerontol 43: males aging symptoms, health and Pols HA, Lamberts SW, Grobbee
M163M169 well-being in elderly community- DE (2003) Endogenous hormones
Thompson IM, Goodman PJ, Tangen dwelling men is not related to and carotid atherosclerosis in
CM, Lucia MS, Miller GJ, Ford LG, circulating androgen levels. elderly men. Am J Epidemiol
Lieber MM, Cespedes RD, Atkins Psychoneuroendocrinology 157:2531
JN, Lippman SM, Carlin SM, Ryan 29:201214 Vandenberghe G, Weekers F, Baxter
A, Szczepanek CM, Crowley JJ, TSjoen GG, Giagulli VA, Delva H, RC, Wouters P, Iranmanesh A,
Coltman CA Jr (2003) The Crabbe P, De Bacquer D, Kaufman Bouillon R, Veldhuis JD (2001)
influence of finasteride on the JM (2005a) Comparative assessment Five-day pulsatile gonadotropin
development of prostate cancer. in young and elderly men of the releasing hormone administration
N Engl J Med 349:215224 gonadotropin response to unveils combined hypothalamic-

369
Chapter 16: Androgens in male senescence

pituitary-gonadal defects underlying Iranmanesh A, Bolton WK (1993) Metabolism in male senescence.


profound hypoandrogenism in men Evidence for attenuation of J Clin Endocrinol Metab 34:
with prolonged critical illness. J Clin hypothalamic gonadotropin- 730735
Endocrinol Metab 86:32173226 releasing hormone (GnRH) impulse Vermeulen A, Giagulli VA, De
Van Den Saffele JK, Goemaere S, De strength with preservation of GnRH Schepper P, Buntinx A, Stoner E
Bacquer D, Kaufman JM (1999) pulse frequency in men with (1989a) Hormonal effects of an
Serum leptin levels in healthy ageing chronic renal failure. J Clin orally active 4-azasteroid inhibitor
men: are decreased serum Endocrinol Metab 76:648654 of 5 alpha-reductase in humans.
testosterone and increased adiposity Veldhuis JD, Iranmanesh A, Godschalk Prostate 14:4553
in elderly men the consequence of M, Mulligan T (2000) Older men
Vermeulen A, Deslypere JP, Kaufman
leptin deficiency? Clin Endocrinol manifest multifold synchrony
JM (1989b) Influence of antiopioids
51:8188 disruption of reproductive
on luteinizing hormone pulsatility
Van Pottelbergh I, Lumbroso S, neurohormone outflow. J Clin
in aging men. J Clin Endocrinol
Goemaere S, Sultan C, Kaufman JM Endocrinol Metab 85:14771486 Metab 68:6872
(2001) Lack of influence of the Veldhuis JD, Zwart A, Mulligan T,
Vermeulen A, Kaufman JM, Giagulli
androgen receptor gene, CAG Iranmanesh A (2001) Muting of
VA (1996) Influence of some
repeat polymorphism on sex steroid androgen negative feedback unveils
biological indices on sex hormone
status and bone metabolism in impoverished gonadotropin-
binding globulin and androgen
elderly men. Clin Endocrinol releasing hormone/luteinizing
levels in aging and obese males.
55:659666 hormone secretory reactivity in
J Clin Endocrinol Metab 81:
Van Pottelbergh I, Goemaere S, healthy older men. J Clin Endocrinol
18211827
Kaufman JM (2003a) Bioavailable Metab 86:529535
Vermeulen A, Goemaere S, Kaufman
estradiol and an aromatase gene Vermeulen A (1991) Clinical Review
JM (1999a) Sex hormones, body
polymorphism are determinants of 24: Androgens in the aging male.
composition and aging. Aging Male
bone mineral density changes in J Clin Endocrinol Metab 73:221224
2:816
men over 70 years of age. J Clin Vermeulen A (2001) Androgen
Endocrinol Metab 88:30753081 replacement therapy in the aging
Vermeulen A, Verdonck L, Kaufman
JM (1999b) A critical evaluation of
Van Pottelbergh I, Braeckman L, De male a critical evaluation. J Clin
simple methods for the estimation
Bacquer D, De Backer G, Kaufman Endocrinol Metab 86:23802390
of free testosterone in serum. J Clin
JM (2003b) Differential Vermeulen A, Deslypere JP (1986) Endocrinol Metab 84:36663672
contribution of testosterone and Intratesticular unconjugated
estradiol in the determination of steroids in elderly men. J Steroid Wallace EM, Pye SD, Wild ST, Wu
cholesterol and lipoprotein profile Biochem 24:10791083 FCW (1993) Prostate specific
in healthy middle-aged men. antigen and prostate gland size in
Atherosclerosis 166:95102 Vermeulen A, Kaufman JM (1992) men receiving exogenous
Role of the hypothalamo-pituitary testosterone for male contraception.
Veldhuis JD, King JC, Urban RJ, Rogol function in the hypoandrogenism of
AD, Evans WS, Kolp LA, Johnson Int J Androl 16:3540
healthy aging. J Clin Endocrinol
ML (1987) Operating characteristics Metab 74:1226A1226C Wang C, Chan V, Yeung RTT (1978)
of the male hypothalamo-pituitary- Effect of surgical stress on pituitary
gonadal axis. Pulsatile release of Vermeulen A, Kaufman JM (2002) testicular function. Clin Endocrinol
testosterone and follicle stimulating Diagnosis of hypogonadism in the 9:255266
hormone and their temporal aging male. Aging Male 5:170176
Wang C, Nieschlag E, Swerdloff R,
coupling with luteinizing hormone. Vermeulen A, Verdonck G (1992) Behre HM, Hellstrom WJ, Gooren
J Clin Endocrinol Metab 65:929941 Representativeness of a single point LJ, Kaufman JM, Legros JJ,
Veldhuis JD, Urban RJ, Lizarralde G, plasma testosterone level for the Lunenfeld B, Morales A, Morley JE,
Johnson ML, Iranmanesh A (1992) long term hormonal milieu in men. Schulman C, Thompson IM,
Attenuation of luteinizing hormone J Clin Endocrinol Metab 74:939942 Weidner W, Wu FCW (2008)
secretory burst amplitude as a Vermeulen A, Stoica T, Verdonck L Investigation, treatment and
proximate basis for the (1971) The apparent free monitoring of late-onset
hypoandrogenism of healthy aging testosterone concentration, an index hypogonadism in males. Eur
men. J Clin Endocrinol Metab of androgenicity. J Clin Endocrinol J Endocrinol 159:507514
75:707713 Metab 33:759767 Wheeler MJ, Barnes SC (2008)
Veldhuis JD, Wilkowski MJ, Zwart AD, Vermeulen A, Rubens R, Verdonck L measurement of testosterone in the
Urban RJ, Lizarralde G, (1972) Testosterone secretion and diagnosis of hypogonadism in the

370
Chapter 16: Androgens in male senescence

ageing male. Clin Endocrinol Wu FC, Tajar A, Pye SR, Silman AJ, Yeap BB, Hyde Z, Norman PE, Chubb
69:515525 Finn JD, ONeill TW, Bartfai G, SAP, Golledge J (2010) Associations
Winters SJ, Sherins RJ, Troen P (1984) Casanueva F, Forti G, Giwercman of total testosterone, sex hormone-
The gonadotropin suppressive A, Huhtaniemi IT, Kula K, Punab binding globulin, calculated free
activity of androgen is increased M, Boonen S, Vanderschueren D; testosterone, and luteinizing
in elderly men. Metabolism The European Male Aging Study hormone with prevalence of
33:10521059 Group (2008) Hypothalamic- abdominal aortic aneurysm in older
pituitary-testicular axis disruptions men. J Clin Endocrinol Metab
Winters SJ, Atkinson L for the in older men are differentially 95:11231130
Testoderm Study Group (1997) linked to age and modifiable risk
Serum LH concentrations in Zitzmann M, Nieschlag E (2003) The
factor: the European male aging CAG repeat polymorphism within
hypogonadal men during study. J Clin Endocrinol Metab
transdermal testosterone the androgen receptor gene and
93:27372745 maleness. Int J Androl 26:7683
replacement through scrotal skin:
further evidence that ageing Wu FCW, Tajar A, Beynon JM, Pye SR, Zitzmann M, Brune M, Kornmann B,
enhances testosterone negative Silman AJ, Finn JD, ONeill TW, Gromoll J, Junker R, Nieschlag E
feedback. Clin Endocrinol 47: Bartfai G, Casanueva FF, Forti G, (2001) The CAG repeat
317322 Giwercman A, Han TS, Kula K, polymorphism in the AR gene
Lean MEJ, Pendleton N, Punab M, affects bone density and bone
Winters SJ, Brufsky A, Weissfeld J, Boonen S, Vanderschueren D,
Trump DL, Dyky MA, Hadeed V metabolism in healthy males. Clin
Labrie F, Huhtaniemi IT, for the Endocrinol 55:649657
(2001) Testosterone, sex hormone EMAS Group (2010)
binding globulin and body Identification of late-onset Zitzmann M, Faber S, Nieschlag E
composition in young adult hypogonadism in middle-aged and (2006) Association of specific
African, American and elderly men. N Engl J Med 363: symptoms and metabolic risks
Caucasian men. Metabolism 123135 with serum testosterone in older
50:12421247 men. J Clin Endocrinol Metab
Yeap BB, Chubb SAP, Hyde Z, 91:43354343
Woolf PD, Hamill RW, McDonald JV, Jamrozik K, Hankey GJ, Flicker L,
Lee LA, Kelly M (1985) Transient Norman PE (2009a) Lower serum Zmuda JM, Cauley JA, Kriska A, Glynn
hypogonadotropic hypogonadism testosterone is independently NW, Gutai JP, Kuller LH (1997)
caused by critical illness. J Clin associated with insulin resistance in Longitudinal relation between
Endocrinol Metab 60: non-diabetic older men: the health endogenous testosterone and
444450 in men study. Eur J Endocrinol cardiovascular disease risk factors in
Worstman J, Eagleton LE, Rosner W, 161:591598 middle-aged men: a 13-year follow-
Dufau ML (1987) Mechanism for up of former Multiple Risk Factor
Yeap BB, Hyde Z, Almeida OP, Intervention Trial Participants. Am
the hypotestosteronemia of the sleep Norman PE, Chubb P, Hamrozik K,
apnea syndrome. Am J Med Sci J Epidemiol 146:609617
Flicker L, Hankey GJ (2009b)
293:221225 Lower testosterone levels predict Zumoff B, Bradlow L, Finkelstein J,
Wu FC, von Eckardstein A (2003) incident stroke and transient Boyar RM, Hellman L (1976) The
Androgens and coronary ischemic attack in older men. influence of age and sex on the
artery disease. Endocr Rev 24: J Clin Endocrinol Metab 94: metabolism of testosterone. J Clin
183217 23532359 Endocrinol Metab 42:703706

371
Chapter
Androgen therapy in non-gonadal disease

17 David J. Handelsman

17.1 Introduction 372 17.7 Bone disease 383


17.2 Liver disease 374 17.8 Critical illness, trauma and surgery 384
17.2.1 Cirrhosis 374 17.8.1 Muscle wasting 384
17.2.2 Hepatitis 374 17.8.2 Skin healing 385
17.2.3 Androgen-induced liver disorders 374 17.8.3 Rehabilitation 386
17.3 Hematological disorders 375 17.9 Immune disease 386
17.3.1 Erythropoiesis and polycythemia 375 17.9.1 AIDS/HIV wasting 386
17.3.2 Anemia due to marrow failure 375 17.9.2 HIV without wasting 387
17.3.3 Myeloproliferative disorders 376 17.10 Malignant disease 388
17.3.4 Thrombocytopenia 377 17.11 Respiratory disease 389
17.4 Renal disease 377 17.11.1 Chronic obstructive lung
17.4.1 Effect of androgens on renal disease 389
function 378 17.11.2 Obstructive sleep apnea 390
17.4.2 Anemia of end-stage renal failure 378 17.11.3 Asthma 391
17.4.3 Growth of boys on hemodialysis 380 17.12 Neurological disease 391
17.4.4 Enuresis 380 17.12.1 Cognitive function 391
17.5 Muscular disorders 380 17.12.2 Headache 392
17.5.1 Frailty 380 17.12.3 Depression 392
17.5.2 Muscular dystrophies 380 17.12.4 Other neurological disease 393
17.6 Rheumatological diseases 381 17.13 Vascular disease 393
17.6.1 Hereditary angioedema 381 17.14 Body weight 394
17.6.2 Rheumatoid arthritis 381 17.14.1 Wasting 394
17.6.3 Other rheumatological disorders 17.14.2 Obesity 394
(systemic lupus erythematosus, 17.15 Key messages 395
Raynauds, systemic sclerosis and 17.16 References 395
Sjgrens disease, chronic urticaria) 382

17.1 Introduction with sufficiently severe or prolonged underlying ill-


Male reproductive function is markedly influenced by ness, this secondary androgen deficiency might con-
non-gonadal disease, with mild androgen deficiency tribute to the disease pathophysiology and clinical
almost a universal consequence of chronic disease. manifestations. Furthermore, androgens are thera-
While this centrally mediated hormonal-axis response peutic drugs with potent effects on androgen-sensitive
to chronic illness is inherently reversible, in theory, tissues, such as muscle, bone, brain, liver or adipose

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

372
Chapter 17: Androgen therapy in non-gonadal disease

tissue, which may be exploited pharmacologically for biochemical consequence of systemic disease, which
therapeutic benefit, subject to comparable efficacy, is reversible upon alleviation of the underlying disorder.
safety and cost-effectiveness criteria as applicable to If the androgen deficiency is unusually severe or pro-
other xenobiotic drugs. longed, it might contribute to morbidity from the under-
As an adjunct to standard medical care, androgen lying disease but is unlikely to change the mortality
therapy may be considered as either physiological (Liu et al. 2003a). The latter follows from recognizing
androgen replacement or, more usually, as pharmaco- that even severe, lifelong androgen deficiency has min-
logical androgen therapy. Androgen replacement ther- imal influence on life expectancy, as seen in congenital
apy aims to replicate endogenous androgen exposure, genetic androgen insensitivity (Quigley et al. 1995), cas-
thereby limiting it to the use of testosterone in doses tration before puberty (Nieschlag et al. 1993; Jenkins
intended to produce physiological blood testosterone 1998) or Klinefelter syndrome (Bojesen et al. 2004); by
concentrations. To the degree it replicates endogenous contrast, castration of institutionalized adults has been
androgen exposure, the expectation for safety may reported to prolong (Hamilton and Mestler 1969) or
reasonably be compared with the benchmark of life- shorten (Eyben et al. 2005) life expectancy, although
long health experience of eugonadal men. the latter studies are severely confounded by the reasons
By contrast, pharmacological androgen therapy prompting castration (Eyman et al. 1990). Hence
is no different from pharmacotherapy with any xeno- pharmacological androgen therapy in systemic disease
biotic drug used to achieve a therapeutic goal. It utilizes can realistically aim to palliate symptoms and improve
any androgen, without restriction to testosterone or QoL without expecting to improve mortality unless it
reference to replacement doses, to optimal effect as ameliorates the natural mortality history of the under-
judged by the standards of efficacy, safety and cost- lying disease. Furthermore, the potential short-term
effectiveness applicable to other drugs. Pharmacological benefits of pharmacological androgen therapy on QoL
androgen therapy has usually involved synthetic, orally must be balanced against potentially detrimental long-
active androgens rather than testosterone, because of term effects of androgen therapy.
industry preference for oral medications. Nearly all Recognizing the few well-established indications for
orally active androgens are from the 17a-alkylated class pharmacological androgen therapy, placebo controls
of synthetic androgens which is now obsolete for andro- remain the hallmark of high-quality clinical studies. In
gen replacement therapy due to its class-specific risk of addition, desirable clinical-trial design features include
hepatotoxicity and the availability of suitable alter- reliable, realistic diagnostic criteria, adequate power and
native testosterone products. The development of non- duration, with valid, objective clinical end-points (rather
steroidal androgens, marketed as selective androgen than surrogates). Unfortunately, few studies fulfill such
receptor modulators (SARMs), offers new possibilities stringent criteria, and the available body of knowledge
for adjuvant pharmacological androgen therapy. In therefore provides little reliable guidance for practical
contrast to the full spectrum of androgen effects of therapeutics. Therefore this chapter focuses on better
testosterone, such SARMs would be pure androgens controlled clinical studies rather than covering the pleth-
not subject to tissue-specific activation by aromatiza- ora of papers reported over more than seven decades
tion to a corresponding estrogen or to amplification of since testosterone was first used clinically (Hamilton
androgenic potency by 5a-reduction. In this context the 1937). A comprehensive account of early, mostly poorly
endogenous pure androgens nandrolone and DHT can or uncontrolled, studies of androgen therapy up to the
be considered prototype SARMs. SARMs are not the mid-1970s is contained in classical comprehensive text-
modern embodiment of so-called anabolic steroids, books (Krskemper 1968; Kochakian 1976).
an outdated term referring to hypothetical but non- Effects of androgen replacement therapy (Chapter
existent non-virilizing androgens targeted exclusively 14), as well as androgen effects on bone (Chapter 8),
to muscle, a failed concept lacking biological proof of cardiovascular disease (Chapter 10) and related
principle (Handelsman 2011). metabolic disorders (Chapter 11), prostate diseases
The goals of androgen therapy for non-gonadal dis- (Chapter 13) and male aging (Chapter 16), as well
ease must be considered in relation to the natural history as in women (Chapter 23), are covered in detail
of the underlying disease. Mild androgen deficiency, elsewhere in this book. Observational studies of sys-
evident in altered blood hormonal levels but without temic disease effects on male reproductive health are
distinctive clinical features, is a universal, non-specific reviewed elsewhere (Handelsman 2010a).

373
Chapter 17: Androgen therapy in non-gonadal disease

17.2 Liver disease group receiving placebo without dietary supplemen-


tation (Mendenhall et al. 1993). This study showed
17.2.1 Cirrhosis no overall survival benefit on primary intention-to-
In studies dating back to the 1960s, pharmacological treat analysis; however, post-hoc subgroup analysis
androgen therapy does not alter the natural history claiming a significant short-term survival benefit
of alcoholic cirrhosis (Rambaldi and Gluud 2006). remained unconvincing due to the study design
The earliest controlled studies of androgen therapy and analysis methodology. Another small random-
to ameliorate the natural history of alcoholic cirrhosis ized, controlled study of 19 men and 20 women with
claimed a survival benefit (Wells 1960), whereas alcoholic hepatitis treated with 80 mg oxandrolone,
another failed to confirm these findings (Fenster parenteral nutrition, both or neither for 21 days
1966); however, neither was large nor long enough demonstrated modest improvement in hepatic bio-
to be definitive. The best evidence derived from the chemical function but did not report other clinical
Copenhagen Study Group for Liver Disease, which end-points (Bonkovsky et al. 1991).
enrolled 221 men with alcoholic cirrhosis in a three-
year prospective, double-blinded, randomized, placebo-
controlled study, testing oral micronized testosterone 17.2.3 Androgen-induced liver disorders
(600 mg daily) (Copenhagen Study Group for Liver A consistent adverse feature of pharmacological
Diseases 1986). This study showed convincingly no androgen therapy, regardless of indication, is the
benefit in mortality, hepatic histology, liver hemody- risk of 17a-alkylated androgen-induced liver dis-
namics and biochemical function or in sexual dysfunc- orders (Ishak and Zimmerman 1987). These involve
tion. The negative outcome with sufficient power to biochemical effects on hepatic function, hepatotoxi-
exclude a 35% decrease in mortality was at variance city (hepatitic or cholestatic) and liver tumor devel-
with previous poorly controlled reports (Kopera 1976). opment (benign or malignant), and peliosis hepatis,
The observation of portal vein thrombosis in three men reflecting the full range of class-specific adverse
treated with testosterone may be related to the dis- effects of oral 17a-alkylated androgens. No reliable
tinctive testosterone pharmacokinetics in chronic estimates of the incidence or prevalence of such
liver disease (Nieschlag et al. 1977; Gluud et al. alkylated androgen-induced hepatotoxicity are
1981), demonstrating markedly supraphysiological available, although claims based on post-marketing
peripheral blood testosterone concentrations at stand- surveillance suggest a low prevalence with low doses
ard clinical doses, presumably due to the high blood used in women in some (Phillips et al. 2003; Orr and
SHBG levels in chronic liver disease, and reflecting Fiatarone Singh 2004) but not all reviews (Velazquez
even more extreme portal blood testosterone concen- and Alter 2004). The East German national sports
trations(Gluud et al. 1987; 1988). doping program using oral 17a-alkylated androgens
resulted in deaths from liver failure and chronic
liver disease (Franke and Berendonk 1997). Every
17.2.2 Hepatitis marketed 17a-alkylated androgen is associated with
Short-term controlled studies of androgen therapy in hepatotoxicity; whereas other androgens (1-methyl
men with alcoholic hepatitis provide no convincing androgens, nandrolone, testosterone, DHT) have
evidence of any clinical benefit (Rambaldi and Gluud minimal risk (Ishak and Zimmerman 1987; Velaz-
2006). A prospective, randomized, multicenter quez and Alter 2004). Biochemical hepatic function
Veterans Administration study claimed a mortality is consistently impaired by oral 17a-alkylated andro-
benefit after 30 days of oxandrolone treatment gens, most frequently manifest clinically as cholesta-
(80 mg daily), compared with placebo in 263 men sis. Androgen-induced hepatitis, peliosis and tumors
presenting with alcoholic hepatitis (Mendenhall are less frequent but unpredictable. Blood SHBG
et al. 1984). The imprecise entry criteria and end- concentrations are significantly reduced by any
points (Maddrey 1986) and short-term benefits of oral androgen as well as supraphysiological circulat-
that study were not confirmed in a further study ing testosterone concentrations in peripheral or
of 271 poorly nourished men with alcoholic hepatitis portal blood (Conway et al. 1988). This indicates
randomized to treatment with oxandrolone plus that SHBG can serve as a useful, sensitive index of
high calorie food supplements compared with a hepatic androgen over-dosage.

374
Chapter 17: Androgen therapy in non-gonadal disease

17.3 Hematological disorders angina or transient ischemic attack) may warrant


venesection. Following recovery from testosterone-
17.3.1 Erythropoiesis and polycythemia induced polycythemia, its dose dependency allows
Androgen therapy has long been used clinically for resuming testosterone treatment with careful
to stimulate erythropoiesis (Shahidi 1973), since the monitoring, using a steady-state preparation gel or
original observational study of 68 women with breast long-acting depot.
cancer demonstrating significant, sometimes dra-
matic, increases in hemoglobin after administering
100 mg testosterone or dihydrotestosterone propion- 17.3.2 Anemia due to marrow failure
ate injections three times weekly (Kennedy and In severe aplastic anemia, a major study of 110
Gilbertsen 1957). In addition, androgen therapy has patients compared HLA-identical marrow transplant-
smaller and less consistent effects on other bone ation with oral, im or no androgen therapy (Camitta
marrow cell lineages that produce neutrophils and et al. 1979). This showed a major survival advantage
platelets. Androgen therapy increases hemoglobin in (70% vs. 35% six-month survival) for 47 patients
healthy men (Palacios et al. 1983; Wu et al. 1996) as having HLA-identical bone marrow transplantation
well as augmenting the hemoglobin responses to compared with 63 patients in whom no donor was
recombinant human erythropoietin (EPO) in renal available who were randomized to oral (oxymetho-
anemia (Ballal et al. 1991) and iron supplementation lone 35 mg/kgday), intramuscular (nandrolone
in iron deficiency anemia (Victor et al. 1967). The decanoate 35 mg/kgwk) or no androgen therapy
molecular mechanism of androgen-induced stimula- (Camitta et al. 1979). The latter three groups did not
tion of erythrogenesis remains poorly understood. differ in survival; a finding consistent with another
Stimulation of erythropoietin secretion, augmenta- small randomized study that showed no survival
tion of erythropoietin action and reduction in circu- benefit due to androgen therapy (50100 mg nandro-
lating levels of hepcidin, an iron transport protein lone phenylpropionate weekly) compared with pla-
(Bachman et al. 2010), have been proposed as non- cebo vehicle injections (Branda et al. 1977).
exclusive mechanisms of action. The hemoglobin In standard non-transplantation treatment for
response to standard testosterone doses is usually aplastic anemia, a randomized crossover study of
modest in magnitude (typically 10 g/l), with overt 44 patients concluded that anti-thymocyte globulin
polycthemia occurring in a small proportion ( 1%) (ATG) was superior to androgen therapy (nandrolone
of normal (Palacios et al. 1983; Wu et al. 1996) or decanoate 5 mg/kgwk). This conclusion was, how-
hypogonadal (Krauss et al. 1991; Drinka et al. 1995) ever, flawed, as half the patients had failed prior
men; whereas high androgen doses produce higher androgen therapy, thus constituting an entry bias
rates ( 8%) of polycythemia (Idan et al. 2010). This against androgen therapy (Young et al. 1988).
risk of androgen-induced polycythemia is increased Coupled with ATG, androgen therapy appears to
in men with higher baseline hemoglobin (Idan et al. offer morbidity but not mortality benefit in aplastic
2010), on higher androgen dose (Idan et al. 2010; anemia. A randomized, controlled multicenter study
Ip et al. 2010) or using injectable testosterone esters of the European Bone Marrow Transplantation in
(Jockenhvel et al. 1997; Siddique et al. 2004) rather Severe Aplastic Anaemia Study Group of 134 patients
than more stable, steady-state testosterone delivery with newly diagnosed severe aplastic anemia receiving
transdermally (Lakshman and Basaria 2009) or by standard therapy (including ATG and methylpredni-
depot implants (Handelsman et al. 1997); while the solone) demonstrated an improvement in transfusion
suspected contributions of high-altitude residence independence due to treatment with oxymetholone
and hypoxia (Gore et al. 2007) and underlying (2 mg/kgday) compared with placebo (Bacigalupo
chronic respiratory failure (Kent et al. 2011) or sleep et al. 1993). However, there was no overall benefit in
apnea (Choi et al. 2006) remain to be confirmed. survival that was determined principally by the sever-
Androgen-induced polycythemia may be asymptom- ity of disease based on leukocyte count. These find-
atic or produce significant clinical effects due to ings confirmed the benefit of androgen therapy on
hyperviscosity and/or ischemia. It is usually reversible transfusion independence but not survival from two
following interruption of androgen treatment, but smaller randomized, placebo-controlled studies
occasionally acute clinical circumstances (e.g. unstable involving 61 patients using oral metenolone acetate

375
Chapter 17: Androgen therapy in non-gonadal disease

(23 mg/kgday) (Li Bock et al. 1976; Kaltwasser et al. analyses, it remains ultimately difficult to conclude
1988), but contradict another randomized, placebo- whether underlying disease prognosis or drug effects
controlled study which found no benefit from androgen explained the differences in group survival. In a follow-
therapy (fluoxymesterone 25 mg/m2day or oxyme- up study from the same cohort who survived at least
tholone 4 mg/kgday) over placebo in 53 patients two years from initial randomization, 137 patients were
(Champlin et al. 1985). None of these studies reported re-randomized to rapid (3 month) or slow (20 month)
survival benefits or formally evaluated QoL, and all withdrawal of their original androgen therapy. The
frequently observed female virilization. slow withdrawal group had a higher rate of maintained
An important pair of studies attempted to define remission consistent with androgen therapy, having
the optimal dosage and type of androgen therapy for maintained a clinical benefit, presumably via main-
aplastic anemia (French Cooperative Group for the tenance of hemoglobin levels, but no survival data
Study of Aplastic and Refractory Anaemias 1986). In were reported (Najean and Joint Group for the Study
the first study, 110 patients were randomized into of Aplastic and Refractory Anaemias 1981). More
four groups according to androgen (norethandrolone, recent findings continue to support an effective role
fluoxymesterone) and dose (high, 1 mg/kgday; low, for androgen therapy as part of immunosuppressive
0.2 mg/kgday). Survival was mainly influenced by (non-transplant) therapy for aplastic anemia (Leleu
disease severity, but, in less severe cases, high-dose et al. 2006).
androgen therapy significantly improved survival Overall, androgen therapy does not improve sur-
over low-dose androgen therapy. Despite randomiza- vival in aplastic anemia but provides a morbidity
tion, there were imbalances between treatment groups benefit by maintaining hemoglobin and transfusion
with respect to disease severity and age that under- independence, although the improved quality of life
mine the interpretability of the findings. In the second has not been quantified. In severe aplastic anemia,
study, 125 patients were randomized to four different bone marrow transplantation from an HLA-identical
androgens norethandrolone, stanozolol, fluoxymes- sibling (if feasible) is the preferred treatment and
terone (all at 1 mg/kgday) or testosterone undecano- superior to androgen therapy. Androgen therapy
ate (1.7 mg/kgday). The fluoxymesterone treatment may be useful in less severe aplastic anemia for which
group had the best and stanozolol the worst survival; bone marrow transplantation is not available or justi-
with norethandrolone and testosterone undecanoate fied (Marwaha et al. 2004; Jaime-Perez et al. 2011), or
being equivalent and intermediate in efficacy. Once where the primary disease is refractory or relapses
again, however, the treatment groups were unbal- after primary marrow transplantation (Freder and
anced with respect to disease severity and age. Hence Valent 2011). However, the relative merits of andro-
the reported benefit limited to the less severe and gen therapy compared with HLA-non-identical bone
older (>30 year) cases remains dubious. The super- marrow transplantation or in the presence of failing
iority of any specific androgen remains to be or failed bone marrow transplantation have not been
unequivocally demonstrated, with particular difficulty clearly defined. Although it is prudent to avoid inject-
in comparing effective doses of different androgens. able androgens in a population that may be thrombo-
The French Cooperative Study Group also reported cytopenic, the preponderant use of oral 17a-alkylated
a series of cohort studies examining the efficacy of androgens in aplastic anemia appears unjustified
androgen therapy in patients with aplastic anemia. when non-hepatotoxic oral androgens such as
Their initial cohort randomized 352 men and women 1-methyl androgens (metenolone, mesterolone) and
to treatment with metandienone (1 mg/kgday), testosterone undecanoate appear to be equally effective.
oxymetholone (2.5 mg/kgday), metenolone acetate
(2.5 mg/kgday) or norethandrolone (1 mg/kgday).
The metandienone group had the best, whereas oxy- 17.3.3 Myeloproliferative disorders
metholone and metenolone groups exhibited equally The use of androgen therapy in other causes of bone
the worst two-year survival from randomization marrow failure has been less extensively studied. One
(Cooperative Group for the Study of Aplastic and controlled study of 29 patients with myeloprolifera-
Refractory Anaemias 1979). However, treatment tive disorders randomized patients to treatment with
groups were unbalanced for disease severity, the princi- fluoxymesterone (30 mg daily) compared with trans-
pal determinant of survival. Despite post-hoc stratified fusions alone, but was terminated prematurely due to

376
Chapter 17: Androgen therapy in non-gonadal disease

slow recruitment and poor hemoglobin response, end-stage renal disease, during dialysis or after renal
with only 4/14 achieving an increase of >10 g/l transplantation. Only a single randomized, controlled
(Brubaker et al. 1982). These findings are supported study has examined androgen replacement therapy in
by another randomized study of 56 patients with uremic men (van Coevorden et al. 1986). Nineteen
myelodysplasia which found oral metenolone acetate regularly hemodialyzed men were randomized to
(2.5 mg/kgday) no better than intravenous cytarabine receive either oral testosterone undecanoate (240 mg
or symptomatic maintenance therapy (Najean and daily) or placebo for 12 weeks. Although libido and
Pecking 1979). Androgens remain part of standard sexual activity increased, hemoglobin was unchanged
supportive care for myeloproliferative disorders and no other androgen effects on bone, muscle, cog-
(Reilly 2006). nition and well-being were reported. Although trans-
dermal testosterone has similar pharmacokinetics in
17.3.4 Thrombocytopenia uremic as in hypogonadal men (Singh et al. 2001), a
phase IV single-center study showed that daily
Experimental evidence suggests a role for endo-
administration of 100 mg transdermal 1% gel failed
genous (Whitnall et al. 2000) or synthetic androgens
to consistently increase serum testosterone or DHT in
(Hosseinimehr et al. 2006) to reduce irradiation-
40 uremic men with reduced blood testosterone levels
induced marrow failure of platelets or leukocytes.
(Brockenbrough et al. 2006). Consequently the failure
A beneficial effect of androgen therapy in thrombo-
to modify EPO requirement or clinical features
cytopenia due to marrow failure has been suggested
(hemoglobin, bone density, body composition, lipids,
by a study in myelodysplasia associated with thrombo-
sexual function or mood) remain inconclusive, and
cytopenia, in which 20 patients were randomized to
further studies achieving an increase in blood testos-
receive either danazol (600 mg daily) or fluoxymester-
terone are required. A Cochrane review of therapeutic
one (1 mg/kgday). Although both groups had an
options for improving sexual function in chronic
impressive response in termination of clinical bleeding
kidney disease failed to evaluate the effects of andro-
(6/6) and increasing platelet count (11/20), the lack of a
gens (Vecchio et al. 2010).
placebo group means that the contribution of natural
Pharmacological androgen therapy has been evalu-
remission could not be evaluated (Wattel et al. 1994).
ated in a randomized, placebo-controlled trial of nan-
The role of androgen therapy in immune throm-
drolone decanoate in dialyzed patients (Johansen et al.
bocytopenic purpura (ITP) remains poorly defined in
1999). Twenty-nine patients were randomized by
the absence of controlled clinical trials. Two short-
sequential allocation to 100 mg nandrolone decanoate
term observational studies have reported that danazol
intramuscularly each week (n 14) or saline placebo
increases platelet counts in ITP as well as decreasing
(n 15) for six months. Lean body mass (measured by
prednisone requirement (Ambriz et al. 1986) and
DXA), timed walking and stair-climbing speed were all
reducing platelet-reactive immunoglobulin G (Ahn
increased; self-reported fatigue fell but there was no
et al. 1983). Reports of long-term usage indicate sus-
change in handgrip strength. Peak oxygen consump-
tained remission in over half the patients but with
tion was also increased at three months, but not sig-
significant adverse hepatic and virilization effects
nificantly so by the end of the sixth month. Larger
(Maloisel et al. 2004; Rice 2004).
placebo-controlled clinical studies of longer duration
are needed to determine whether the impressive short-
17.4 Renal disease term benefits are sustainable and/or improve survival.
Men with chronic renal failure exhibit many non-specific A well-controlled, randomized phase II dose-finding
features consistent with androgen deficiency including study of 54 patients on optimized EPO therapy showed
gynecomastia, impotence, testicular atrophy, impaired that 24 weeks of treatment with doses of 50, 100 or 200
spermatogenesis and infertility, as well as somatic dis- mg nandrolone decanoate weekly (halved in women),
orders of bone, muscle and other androgen-responsive for 24 weeks increased appendicular lean mass in a
tissues (Handelsman 1985; Handelsman and Dong 1993; dose-dependent manner (Macdonald et al. 2007). This
Handelsman and Liu 1998). Such consequences of occurred without increasing fluid overload or any con-
uremia are partially rectified during dialysis but reversed sistent improvement in physical functioning, but vir-
only by renal transplantation. There is little information ilization of women was intolerable at doses higher than
on androgen replacement therapy in patients with 50 mg weekly. It was concluded that body composition

377
Chapter 17: Androgen therapy in non-gonadal disease

is enhanced by doses of 200 mg for men (50 mg for pressure and reducing renal function. Hence the
women) weekly, but improving physical functioning potential amelioration due to the age-related decline
may require additional exercise interventions. in circulating androgen levels may represent an unrec-
ognized benefit (Reckelhoff et al. 2005).
17.4.1 Effect of androgens on renal
function 17.4.2 Anemia of end-stage renal failure
Based on the renotrophic effects of androgens The anemia of end-stage renal failure has multiple
(Mooradian et al. 1987), it has long been speculated contributory factors including EPO deficiency, toxic
that androgen therapy in patients with chronic renal inhibitors of EPO action, androgen deficiency, micro-
failure or nephrotic syndrome might improve or nutrient deficiency (iron, folate, pyridoxine), blood
slow the deterioration in underlying renal function. loss and hemolysis (Neff et al. 1985). The effect of
However, clinical evidence for renotrophic effects of androgen therapy on hemoglobin involves both
androgen therapy has remained ambiguous due to the increased circulating EPO concentration (Buchwald
lack of adequately powered, placebo-controlled stud- et al. 1977), and augmentation of EPO action (Ballal
ies (Krskemper 1968; Kopera 1976). The best clinical et al. 1991). Erythropoietin deficiency is a major
evidence is derived from a placebo-controlled study of factor (Winearls 1995), and androgen therapy prob-
elderly patients without renal disease (Dontas et al. ably acts mainly by increasing EPO, since androgen
1967) and another evaluating uremic patients but therapy has no effect on hemoglobin after bilateral
without a control group (Wilkey et al. 1960). In nephrectomy (von Hartitzsch and Kerr 1976), when
the first study (Dontas et al. 1967), indices of both the major source of endogenous EPO is removed.
glomerular and tubular function improved with nan- Androgen therapy has consistent effects on EPO
drolone phenylpropionate (25 mg injections weekly) secretion and hemoglobin concentrations (Navarro
after 40 weeks. In the other (Wilkey et al. 1960), and Mora 2001), although circulating EPO is not
well-being and biochemical tests of renal function consistently related to resultant increases in hemo-
improved, but no detailed findings or analysis were globin (Teruel et al. 1995). Endogenous testosterone
presented for the 88 uremic patients treated with is an important physiological determinant of red cell
various doses of injectable testosterone propionate mass in men, since blockade of androgen action
(50 mg daily) or cypionate (100 mg daily to monthly) lowers hemoglobin levels (Weber et al. 1991; Teruel
and oral fluoxymesterone (5 mg daily) (Wilkey et al. et al. 1997). Furthermore, post-transplant erythrocy-
1960). Despite the biological basis for renotrophic tosis may depend on EPO and possibly also endogen-
effects, the lack of adequate clinical evidence pre- ous testosterone (Chan et al. 1992). These findings
cludes an established role for androgen therapy in suggest that androgens also have important actions
the management of chronic renal failure. in augmenting EPO effects. The intertwined roles
More recently, the possibility that androgen ther- of androgen and EPO deficiency and sensitivity in
apy may be detrimental to the function of kidney chronic renal failure remain difficult to disentangle
transplants was suggested based on rodent experi- (Silverberg et al. 2004; Daniell 2006; Diskin 2007).
ments in which androgen therapy hastened, and Two randomized, placebo-controlled studies have
androgen blockade delayed, chronic allograft neph- shown that nandrolone treatment increases hemo-
ropathy (Muller et al. 1999; Antus et al. 2001). These globin in patients with end-stage renal failure. One
effects did not require aromatization of testosterone, randomized 21 men to nandrolone (100 mg weekly)
nor were they gender specific (Antus et al. 2002). or placebo vehicle injections for five months in a
Although no systematic clinical data have been crossover design (Hendler et al. 1974); while another
reported, case reports raised concerns about use of randomized 18 patients to nandrolone decanoate
androgens in patients with kidney and other trans- (200 mg weekly) for three months (Williams et al.
plants (Schofield et al. 2002) and warrant further 1974). Both found significant increases in mean
evaluation. Analogous but wider concerns have been hemoglobin (15 g/l and 10 g/l, respectively), and one
raised about androgen therapy, based on experimental reported a clinically significant decreased transfusion
findings in animal models that androgen may produce requirement (Hendler et al. 1974). A further study
deleterious effects on the kidneys, such as raising blood confirmed the beneficial effects of nandrolone decanoate

378
Chapter 17: Androgen therapy in non-gonadal disease

(200 mg weekly) compared with placebo vehicle for six months) was equivalent to EPO (initial dose
injections for four months (Buchwald et al. 1977); 50 units/kgweek, titrated to maintain hemoglobin
whereas three smaller and less well-conducted between 11 and 13 g/dl) in maintaining hemoglobin
studies failed to show an increase in hemoglobin (Navarro et al. 2002). These studies together suggest
(Li Bock et al. 1976; Naik et al. 1978; van Coevorden that im nandrolone decanoate (200 mg/week) in
et al. 1986). A further randomized, controlled dialyzed men over 50 years of age is as effective
clinical study compared four androgen regimens in as EPO in maintaining hemoglobin. However, the
dialyzed patients, finding that testosterone enanthate relative safety of these treatments requires further
(4 mg/kgwk) and nandrolone decanoate (3 mg/kgwk) clarification.
were more effective in increasing hematocrit than Androgen therapy may also have an adjunctive
oxymetholone (1 mg/kgday) and fluoxymesterone role to EPO, perhaps as an EPO-sparing agent. This
(0.4 mg/kgday). However, whether these differences has been examined in two randomized (Berns et al.
reflected different effective androgen doses, the 1992; Gaughan et al. 1997) and one non-randomized
androgen class (17a-alkylated or not) or route of (Ballal et al. 1991) EPO-controlled studies. In the
administration (including pharmacokinetics) remains most powerful study (Gaughan et al. 1997), 19 dia-
unclear (Neff et al. 1981). Future studies examining lyzed patients were randomized to receive nandrolone
androgen replacement therapy using transdermal tes- (100 mg weekly) plus EPO (4500 U/wk) or EPO alone
tosterone will be of interest, and preliminary studies for 26 weeks. The addition of nandrolone to low-dose
indicate similar pharmacokinetic profile in uremic as EPO (approximately equal to 60 U/kgwk) resulted
in hypogonadal men (Singh et al. 2001). in a significantly greater rise in hematocrit. Similar
There is accumulating evidence that androgen or significant additional increases in hemoglobin were
EPO therapies are equally effective in maintaining reported in a small non-randomized study of eight
hemoglobin in patients with chronic renal disease. men choosing to receive nandrolone decanoate (100
A retrospective analysis of 84 patients receiving andro- mg weekly) plus intermediate dose EPO (6000 U/wk)
gen therapy (nandrolone decanoate 200 mg weekly) compared with EPO alone (Ballal et al. 1991) for
for six months reported that men over 55 years of age 12 weeks. To the contrary, another small but
had the best hemoglobin responses, and that this randomized study employing a higher dose of EPO
response was comparable with those treated with (120 U/kgwk) was unable to detect any benefit of
EPO (Teruel et al. 1996a). This was subsequently nandrolone decanoate (2 mg/kgwk) for 16 weeks plus
confirmed in two controlled, prospective studies EPO compared with the same dose of EPO alone in
which both treated with nandrolone decanoate 12 dialyzed patients (Berns et al. 1992). Whether these
200 mg/wk or EPO (6000 U/wk) for six months. The discrepancies are due to study design, age or EPO
first prospective study found very similar hemoglobin dose remains to be clarified; although it is possible
responses and safety profiles for 18 men over 50 years that androgens have greatest synergism with sub-
of age treated with androgen (nandrolone decanoate maximal EPO dosage, and that the higher EPO dose
200 mg/wk) compared with 6 men under 50 years and obviates any additional androgen-induced increase
16 women receiving EPO (6000 U/wk); however, the in hemoglobin. Randomized prospective studies to
lack of randomization and non-comparability of examine the use of low-dose subcutaneous EPO with
groups by age and gender limits the interpretation of adjunctive androgen therapy are needed (Horl 1999),
these findings (Teruel et al. 1996b). The second study particularly in older men.
(Gascon et al. 1999) randomized 33 patients over A caveat on androgen therapy is the risk of poly-
the age of 65 to receive im nandrolone decanoate cythemia, which occurs as a rare idiosyncratic reac-
200 mg/wk (n 14) or to continue EPO (mean dose tion among men with normal renal function receiving
of 6000 units/week; n 19) for six months, and found exogenous testosterone (Drinka et al. 1995). Testos-
comparable hematological parameters by the end of terone-induced polycythemia may be more common
the study. However, it appears that all seven women among older men receiving im testosterone injections
were allocated, rather than randomized, to EPO since (Hajjar et al. 1997) and less common with more
no woman received nandrolone. Recently, a random- steady-state depot testosterone delivery, but has been
ized, controlled study in 27 men aged over 50 years observed with all forms of exogenous androgen
reported that nandrolone decanoate (200 mg weekly (Jockenhvel et al. 1997).

379
Chapter 17: Androgen therapy in non-gonadal disease

17.4.3 Growth of boys on hemodialysis mental function of aging people; notably the increased
vulnerability to further deterioration, together with
One small, double-blind, placebo-controlled crossover
reduced recuperative power responding to adverse
study examined the effects of testosterone on short-
effects which have relatively minor detrimental
term growth in boys with short stature on hemodialy-
impact at younger age. Although progressive declines
sis (Kassmann et al. 1992). After an eight-week run-in,
in multiple organ systems all contribute to frailty, this
eight boys (mean 3.9 SD below mean height for age)
syndrome is most evident in muscular function. The
on regular hemodialysis were randomized to start on
increased risk of fall, fractures and related adverse
one of two four-week treatment periods separated by
effects among the aging are ascribed to frailty. Conse-
a six-week wash-out period before crossing over to the
quently, frailty has steadily evolved into a framework
other treatment. Treatment consisted of 2 g/m2day
for considering hormonal treatment for aging
of a transdermal gel, corresponding to a topical
(Villareal and Morley 1994) including the use of tes-
daily dose of 50 mg/m2day testosterone or placebo.
tosterone (the andropause hypothesis). Currently,
Although a significant increase in short-term growth
however, there is no evidence that frailty is improved
velocity (using knemometry) was reported overall,
by androgen administration to older men (Sullivan
gain of final height was not reported and cannot be
et al. 2005; Muller et al. 2006; Srinivas-Shankar et al.
predicted from growth velocity. Furthermore, the
2010; Kenny et al. 2010; Atkinson et al. 2010). This
small sample size and unbalanced randomization
may reflect the fact that, although testosterone admin-
were limitations. Further larger and longer studies
istration to older men consistently improves muscle
would be needed before even low-dose androgen ther-
mass and strength, such treatment effects may be
apy could be considered effective or safe.
insufficient to provide clinically meaningful improve-
ment in physical performance or resilience of older
17.4.4 Enuresis men (OConnell et al. 2011).
Following suggestions from the 1940s that androgen
therapy might improve childhood enuresis, a recent
controlled clinical trial involving 30 boys aged 610 17.5.2 Muscular dystrophies
years has claimed a benefit for oral mesterolone treat- The effects of androgen therapy on neuromuscular
ment compared with placebo (El-Sadr et al. 1990). This disorders have been best studied by Griggs et al., in
study may have been flawed as the method of random- a series of careful studies of myotonic dystrophy,
ization leading to 20 being treated with mesterolone a genetic myopathy due to a trinucleotide (CTG)
(20 mg daily for two weeks) compared with 10 on pla- repeat mutation in the myotonin (protein kinase)
cebo (vitamin C) was not explained. The statistically gene. Myotonic dystrophy is associated with testicular
significant increase in cystometric bladder capacity in atrophy and biochemical androgen deficiency com-
the mesterolone-treated group was attributable to 6 boys pared with age-matched healthy men or men with
who had dramatic increases; whereas the remainder did other neuromuscular wasting diseases (Griggs et al.
not differ from the 10 placebo-treated boys. Although no 1985), although serum testosterone does not correlate
adverse effects were reported, the well-known potential with extent of muscle wasting. Since life expectancy
hazards of androgen therapy in prepubertal children, in myotonic dystrophy is determined by respiratory
including premature closure of epiphyses and short stat- muscular weakness leading to terminal pneumonia,
ure, precocious sexual maturation and psychological androgen therapy aiming to improve muscular
sequelae, would require detailed safety evaluation before strength might prolong life. To test this hypothesis,
androgen therapy could be considered acceptable for a a randomized, placebo-controlled study was under-
benign functional disorder with favorable natural his- taken in 40 men with myotonic dystrophy who were
tory in otherwise healthy children. treated with either testosterone enanthate (3 mg/kg)
or placebo injections each week for 12 months
17.5 Muscular disorders (Griggs et al. 1989). In a well-designed two-site study,
muscle mass was increased as indicated by creatinine
17.5.1 Frailty excretion and total body potassium, but there was
Frailty is a clinical syndrome with features derived no difference in quantitative measures of manual or
from the natural history of declining physical and respiratory muscle strength. Crucially, the lack of

380
Chapter 17: Androgen therapy in non-gonadal disease

improved pulmonary function implies that mortality been used, danazol has become standard prophylactic
benefits would be unlikely. Androgen therapy may therapy. This followed a randomized, double-blind,
simply increase the mass of dysfunctional muscle. crossover study which showed increased blood
The same investigators also examined the effect of C1-esterase inhibitor concentration together with a
androgen therapy in boys with Duchenne muscular dramatic decrease (94% vs. 2%) in attack-free 28-day
dystrophy in a randomized, placebo-controlled study periods using 600 mg danazol daily compared with
(Fenichel et al. 2001), following encouraging results placebo in 93 courses among nine patients (Gelfand
from an uncontrolled pilot study of 10 boys treated for et al. 1976). Danazol doses are tapered to minimal
three months (Fenichel et al. 1997). Boys 510 years levels that maintain adequate control of attack, and
of age with Duchenne muscular dystrophy (n 51) this dose minimization may explain the anecdotal
were randomly assigned to receive oxandrolone (0.1 impression that such danazol therapy has minimal
mg/kgday) or placebo for six months. Although the effects on male fertility, although quantitative studies
primary end-point (semi-quantitative average muscle have not been reported. Recent studies suggest that
strength score) and timed functional tests of gait were stanozolol (12 mg daily) is about as effective as
not significantly improved, oxandrolone produced danazol (50200 mg daily), but, despite their efficacy,
a significant increase in some post-hoc comparisons hepatotoxicity and female virilization remain prob-
such as quantitative myometry and in upper limb lems (Hosea et al. 1980; Cicardi et al. 1997). While it
muscle strength score. Despite marginal efficacy, oxan- is assumed that the beneficial effects of androgen
drolone was granted FDA orphan drug status for treat- therapy in reducing frequency and severity of angioe-
ment of Duchenne muscular dystrophy in 1997. While dema attacks are only exhibited by 17a-alkylated
it may produce better growth and fewer side-effects androgens, only very limited studies of non-17a-
than high-dose glucocorticoids, which are more effect- alkylated androgens such as nandrolone, 1-methyl
ive but at the cost of more adverse effects including androgens or testosterone (Spaulding 1960) have been
growth retardation and weight gain, the relative merits reported. Since angioedema requires lifelong prophy-
or oxandrolone (or other safer androgens) compared laxis, further studies of non-hepatotoxic androgens
with intermittent, low-dose prednisone (Dubowitz should be undertaken. Noting that androgen effects
et al. 2002) warrant direct evaluation. Similarly, the are not fast enough for acute treatment, it is possible
discrepancy between these findings is puzzling and the oral route of administration may constitute a
the precise role of pharmacological androgen therapy form of liver targeting (via first-pass exposure) for
in other forms of neurogenetic or degenerative neuro- high hepatic androgen doses, which might not be
muscular disorders also deserves evaluation. feasible or safe for parenteral administration.
Long-term experience among allergy specialists
17.6 Rheumatological diseases continue to show androgens exhibiting reasonable
cost-effectiveness in reducing frequency and severity
17.6.1 Hereditary angioedema of attacks balanced against adverse effects including
The efficacy of oral 17a-alkylated androgens in her- hepatotoxicity and virilization (Sloane et al. 2007;
editary angioedema was established by a small, Bork et al. 2008; Banerji et al. 2008; Craig 2008; Fust
double-blind, placebo-controlled randomized cross- et al. 2011), and androgens remain the most widely
over study (Spaulding 1960) in which six members used prophylaxis (Riedl et al. 2011).
of a single family received multiple periods of treat-
ment or placebo. This study clearly demonstrated the
efficacy of oral methyltestosterone in reducing the 17.6.2 Rheumatoid arthritis
frequency of attacks well before the disease pathogen- The rationale for androgen therapy in rheumatoid
esis was understood. Subsequent studies confirmed arthritis is that (1) the lower prevalence in men sug-
these observations showing that androgen therapy gests a protective role for androgens; (2) active disease
increases C1-esterase inhibitor concentration, par- is associated with reduction in endogenous testosterone
tially rectifying the underlying biochemical deficiency production; (3) androgen effects on muscle and bone
responsible for the disorder (Sheffer et al. 1977). may improve morbidity in rheumatoid arthritis; and
Although other 17a-alkylated oral androgens such as (4) androgen effects (e.g. fibrinolysis) may reduce dis-
fluoxymesterone, oxymetholone and stanozolol have ease activity.

381
Chapter 17: Androgen therapy in non-gonadal disease

The best designed and conducted study of andro- or placebo for nine months (Hall et al. 1996). This
gen therapy involved 107 women with active rheuma- study noted that overall disease activity (defined by
toid arthritis according to American College of biochemical variables and clinical scales) was not
Rheumatology (ACR) criteria on stable standard improved by androgen therapy, and, indeed, signifi-
(steroid, NSAID) treatment for at least three months cantly more men on testosterone therapy experienced
who were randomized to treatment with fortnightly disease flare during the study. The inclusion of men
injections of either androgen therapy (testosterone with inactive rheumatoid arthritis and initial use of an
propionate 50 mg plus progesterone 2.5 mg) or pla- inadequate testosterone dose were limitations of this
cebo for one year (Booij et al. 1996). The inclusion of study. An older double-blind study randomized 40
a very low dose of progesterone, which the authors patients with definite rheumatoid arthritis on stable
claim was biologically ineffective, was based on an old NSAID to treatment with stanozolol 10 mg daily or
clinical practice aiming to reduce virilization from placebo for six months on the basis that androgen
testosterone. Evaluated on a double-blinded, inten- therapy might increase fibrinolysis (Belch et al. 1986).
tion-to-treat basis, this study demonstrated signifi- This study found a significant improvement in the
cant improvement in the erythrocyte sedimentation composite Mallaya disease activity index combining
rate (ESR), pain and disability scores and ACR objective (ESR, hemoglobin, articular scores) and
improvement criteria, but not in the numbers of subjective (pain, morning stiffness) dimensions, des-
tender or swollen joint or joints requiring intra- pite the failure to influence measurable fibrinolysis.
articular steroid injections. There was a high dropout Adverse effects such as hepatotoxicity or virilization
rate (39/107), mostly (28/39) due to inefficacy defined in females were not reported. Whether androgen
as any mid-study increase in anti-rheumatic medica- therapy in men with rheumatoid arthritis can modify
tion; however, these were evenly distributed between the natural history or whether it only improves mood
treatment groups. As expected, virilization was the and toleration of pain and disease remains to be
major adverse effect reported, but there were few clarified.
other side-effects, and tolerability was good, as most
androgen-treated patients (67% vs. 37% on placebo)
wished to continue their allocated medication at the
17.6.3 Other rheumatological disorders
end of the study. The significant benefits of androgen (systemic lupus erythematosus, Raynauds,
therapy over placebo were predominantly in subject- systemic sclerosis and Sjgrens disease,
ive measures rather than objective signs of disease
activity. This raises the possibility that androgen ther- chronic urticaria)
apy may preferentially improve mood or tolerance of Few well-controlled studies of androgen therapy have
disability rather than actually modifying disease been reported in other rheumatological disorders.
impact or natural history. This well-designed study This includes male-preponderant rheumatological
is a model for investigation of pharmacological diseases (ankylosing spondylitis, gout) as well as
androgen therapy in systemic disease. the majority of female-preponderant autoimmune
Other studies of androgen therapy in rheumatoid diseases.
arthritis are small, poorly designed and inconclusive. In systemic lupus erythematosus (SLE), only
One uncontrolled study of seven men with rheuma- two, small, uncontrolled studies (including together
toid arthritis treated with six months of androgen 5 men among 17 patients) using androgen therapy
therapy (oral testosterone undecanoate 120 mg daily) (nandrolone decanoate) have been reported (Hazelton
observed a decline in disease activity (reduced et al. 1983; Lahita et al. 1992). This information is so
numbers of tender joints and analgesic usage) limited that no conclusions can be drawn without
together with minor immunological changes that larger and better-designed studies. Another double-
were not correlated with disease activity. The lack of blind study randomized 28 women with mild to mod-
a placebo group in a disease with a remitting natural erate SLE to treatment with DHEA (200 mg daily) or
history renders such observations unconvincing placebo for three months. Treatment with this weak
(Cutolo et al. 1991). A larger study of 35 men with androgen precursor did not improve SLE disease
definite rheumatoid arthritis randomized them to activity index, number of flares, prednisone usage or
injections of testosterone enanthate (250 mg monthly) physician overall assessment, although there was an

382
Chapter 17: Androgen therapy in non-gonadal disease

improvement in the patients overall assessment of of the underlying disease activity. This is reinforced by
well-being (Van Vollenhoven et al. 1995). These find- well-controlled studies of 60 women with Sjgrens
ings are reinforced by a well-controlled study of 60 syndrome randomized to 200 mg DHEA or placebo
women with quiescent SLE randomized to 200 mg for 12 months, where there was a striking placebo
DHEA or placebo for 12 months, where effects of effect with expectation but not DHEA having clear
placebo and expectation but not of DHEA were apparent benefits (Hartkamp et al. 2008).
observed (Hartkamp et al. 2010). By contrast, another A recent randomized, double-blind study exam-
study of 41 women with glucocorticoid-treated ined the role of stanozolol as an adjunct to standard
SLE reported that the addition of low-dose DHEA antihistamine therapy for chronic urticaria. Patients
(2030 mg) for six months was superior to placebo (20 men, 30 women) were randomized to treatment
for improving QoL (Nordmark et al. 2005). It is not with stanozolol 4 mg daily or placebo in addition to
clear whether this discrepancy reflects differences in antihistamine (cetirizine 10 mg daily) for 12 weeks
underlying SLE activity or the effects of glucocorti- (Parsad et al. 2001). Over 70% improvement in phys-
coid suppression of adrenal function. A study using ician and patient-scored urticaria was observed in
low (female) doses of transdermal testosterone (150 mg 17/26 patients who received stanozolol, but in only
daily) for 12 weeks produced no benefit in 34 women 7/24 patients who received cetirizine alone. This
with SLE (Gordon et al. 2008). highly statistically significant benefit was observed
One study has examined the effects of treatment four weeks after starting treatment and continued
with stanozolol (10 mg daily) or placebo for 24 weeks throughout the study. Whether the benefits of stano-
in primary Raynauds phenomenon and systemic zolol are gender specific, or whether other androgens
sclerosis (Jayson et al. 1991). Although 43 patients are also effective, has not been established.
(19 Raynauds, 24 systemic sclerosis; including only
4 men) entered, only 28 patients (11 Raynauds, 17
systemic sclerosis) completed the study. Compared 17.7 Bone disease
with placebo, stanozolol significantly improved ultra- The role of androgens in bone development and
sonic Doppler index as well as finger pulp and nail disorders is discussed in Chapter 8. Androgen therapy
bed temperatures, but there was no difference in to treat osteoporosis has the advantage for fracture
reported frequency or severity of vasospastic attacks, prevention of not only increasing bone mass but
scleroderma skin score or grip strength. The clinical possessing potentially synergistic beneficial effects
significance of the changes in digital small vessel on muscular strength and mental function to prevent
function recorded in the absence of vasospasm and falls due to frailty: an independent contributor to
without reduction in attack rates is unclear. osteoporotic fractures. The evidence supporting
A double-blind study randomized 20 women with androgen therapy, however, is limited. For treatment
primary Sjgrens syndrome to treatment with andro- of idiopathic osteoporosis, the largest randomized,
gen (nandrolone decanoate 100 mg fortnightly) or pla- placebo-controlled study, involving 327 patients
cebo for six months (Drosos et al. 1988). Androgen treated for nine months with one year of follow-up,
therapy did not produce any significant improvement had inadequate power to detect effects of androgen
over placebo in objective validated measures of xeros- therapy (metandienone 2.5 mg daily) on fracture rates
tomia (stimulated parotid flow rate measurements, (Inkovaara et al. 1983). The only other controlled
labial salivary gland histology), xerophthalmia study randomized 21 men to receive either weekly
(Schirmers I test, slit lamp eye examination after rose injections of nandrolone decanoate 50 mg or no
Bengal staining) or systemic disease (ESR), although treatment for 12 months (Hamdy et al. 1998). It
the subjective assessment of xerostomia by patients and remains unclear whether the inconsistent and transi-
physicians as well as overall patients well-being assess- ent increase in bone density observed was due to the
ment were significantly better on nandrolone. Viriliza- low dose, the minimally aromatized androgen or
tion was reported in nearly all nandrolone-treated small sample size. Additionally, an uncontrolled study
women with this relatively high androgen dose, but has claimed striking increase in lumbar (but not hip)
none discontinued for this reason. Again these studies bone density in non-androgen deficient men treated
reinforce the observations that androgen therapy may with testosterone ester injections 250 mg fortnightly
significantly improve feelings of well-being regardless for six months (Anderson et al. 1997).

383
Chapter 17: Androgen therapy in non-gonadal disease

An important area for androgen therapy to pre- bone and skin recovery where sustained androgen
vent or ameliorate bone loss and fractures may be deficiency is sufficient to contribute significantly to
steroid-induced osteoporosis. High-dose glucocorti- the overall catabolic state. However, the endocrine
coid therapy is commonly used for its immunosup- responses to catabolic states such as critical illness
pressive or anti-inflammatory effects in autoimmune are highly complex, involving widespread dysregula-
and chronic inflammatory diseases and in transplant- tion of all pituitary hormonal axes, so that, if restor-
ation medicine. Two controlled studies have examined ation of any anabolic hormones is effective, it may
androgen therapy in men taking regular high-dose require multiple rather than individual replacement
glucocorticoid treatment. The first reported that tes- (or pharmacological doses) to be effective (Langouche
tosterone may reverse the bone loss due to high-dose and van den Berghe 2006). The success of intensive
glucocorticoid therapy in 15 men with severe asthma insulin therapy to regulate hyperglycemia (van den
(Reid et al. 1996). The subjects were randomly Berghe et al. 2001) has been disputed (Finfer et al.
allocated to monthly testosterone injections (250 mg 2009), leaving uncertainty over targets and outcomes
mixed testosterone esters) or no treatment for 12 in clinical practice (Kavanagh and McCowen 2010),
months, with the control group crossing over to tes- highlighting the caution that, whatever the promise
tosterone treatment for the second 12-month period. of combination pituitary hormonal approaches,
After 12 months of testosterone treatment, lumber rigorous experimental evaluation is required before
spine BMD increased by 5% compared with no adoption (Langouche and van den Berghe 2006). Key
change on placebo. However, no benefit was noted surrogate outcome variables for evaluating the effi-
in bone density overall or in three other sites. The cacy of androgen therapy in such catabolic states
limitations of this study (unblinded, sub-replacement include (1) muscle mass, strength and function;
testosterone dose) are addressed in a larger study (2) bone turnover and wound healing (particularly
randomizing 51 men to fortnightly injections of after burns); as well as (3) health service utilization
testosterone esters 200 mg, nandrolone decanoate variables such as duration of in-hospital stay and rate
200 mg or matching oil vehicle placebo for 12 months and extent of rehabilitation.
(Crawford et al. 2003). This study observed improved
muscular strength with both androgens but improved
lumbar bone density and bone-specific QoL only in 17.8.1 Muscle wasting
men treated with testosterone. This highlights the A number of studies have examined the effects of
importance of aromatization in androgen therapy androgen therapy as an adjunct to elective surgery
for bone but not muscle. Larger studies examining using improved nitrogen balance as a surrogate for
fracture outcome as well as earlier studies aimed muscle mass for their end-point. The best designed
to prevent the rapid initial bone loss would be study randomized 60 patients after colorectal cancer
most valuable. surgery to receive either a single injection of stanozo-
lol (50 mg) or no extra treatment. Participants were
also randomized among three types of post-operative,
17.8 Critical illness, trauma peripheral-vein nutrition (standard dextrose-saline,
and surgery amino acid supplementation or glucose-amino acid-
Critical illness, trauma, burns, surgery and malnutri- fat mixture) and stratified by gender (Hansell et al.
tion all result in a catabolic state characterized by 1989). The primary end-point was cumulative nitro-
acute muscle breakdown which is reversed during gen balance for the first four post-operative days,
recovery. These catabolic states are characteristically and this was consistently and significantly influen-
accompanied by functional hypogonadotropic andro- ced only by nutritional supplementation. Stanozolol
gen deficiency. This is due to functional partial GnRH augmented nitrogen balance only on the third post-
deficiency as pulsatile GnRH administration can operative day in the group receiving amino acid sup-
rescue LH pulsatility and hypoandrogenemia (Aloi plements. This was largely attributable to its effects
et al. 1997; van den Berghe et al. 2001). This has long in women and gave no improvement over standard
led to the hypothesis that androgen therapy might post-operative care on other post-operative days, with
improve mortality or morbidity by pharmacologically other nutritional supplements, nor had any influence on
enhancing nutritional supplementation and muscle, a wide range of other metabolic variables. Importantly,

384
Chapter 17: Androgen therapy in non-gonadal disease

neither convalescence nor complication rates were supplemental treatment with either nandrolone
influenced by androgen or nutritional therapy. decanoate (50 mg) or placebo injections biweekly,
Other studies have largely confirmed these find- aiming to determine whether nitrogen balance could
ings. The first randomized 44 men with tuberculosis be improved within the first 21 days post-operatively
requiring pulmonary resection to treatment with (Lewis et al. 1981). No benefit was observed in nitro-
either high-dose norethandrolone (50 mg daily) or gen balance, weight gain, creatinine output, and
no extra treatment within strata of different inten- serum albumin or immune function. These negative
sity of post-operative hyperalimentation (Webb et al. findings were supported by another study that exam-
1960). This showed a modest, transient effect of ined a higher nandrolone dose. This study random-
androgen therapy on positive nitrogen balance restri- ized 24 patients requiring intravenous alimentation
cted to the first-three post-operative days which was to nandrolone decanoate (100 mg before starting and
absent during the second-three post-operative days. repeated one week later) or no extra treatment, and
The second study randomized 36 patients to one found increased fluid but not nitrogen balance,
injection of stanozolol (50 mg) or placebo one day and did not find any clinical benefits (Young et al.
before surgery, with similar outcomes (Blamey et al. 1983). Another study has examined the use of oral
1984). A third study randomized 30 men after gastric oxandrolone, randomizing 60 patients (including
surgery for duodenal ulcer (vagotomy/pyloroplasty) 5 women) requiring enteral nutrition to oxandrolone
to a single post-operative injection of nandrolone 20 mg each day or placebo for no more than 28 days,
decanoate (50 or 100 mg), parenteral nutrition, both, and reported no differences in nitrogen balance or
or to standard treatment (Tweedle et al. 1973). This clinically relevant outcomes such as infection rate or
study reported that the eight-day post-operative length of stay (Gervasio et al. 2000).
nitrogen balance was best with the combination of
nandrolone plus parenteral nutrition, and that each
alone was superior to standard treatment, but no 17.8.2 Skin healing
clinical outcome measures were reported. The fourth One group of investigators has performed two studies
study randomized 20 patients recovering from mul- examining whether oxandrolone (20 mg/day) can
tiple trauma to receive either nandrolone decanoate promote skin healing after severe burn injury. The
injections (50 mg on day three plus 25 mg on day six) double-blind study randomized 20 patients with
or no extra treatment. It found that nandrolone plus severe burns to receive oxandrolone or matching
standard enteral or parenteral nutrition was superior placebo for at least three weeks commencing two to
to no extra treatment in nitrogen balance, urinary three days after the injury (Demling and Orgill 2000).
3-methyl histidine excretion and amino acid retention Oxandrolone therapy promoted skin healing in the
for the first 10 days of hospitalization (Hausmann standardized donor site, improved nitrogen balance
et al. 1990). The only clinical outcome measure, how- and reduced weight loss, but did not alter length of
ever, was six-month survival, which did not differ hospital stay. Similar results were reported in an
according to androgen therapy. Finally, more recent earlier non-blinded study of 36 patients randomized
studies have shown benefits in burned children ran- to receive GH (n 20) or oxandrolone (n 16)
domized to oxandrolone or placebo. One study (Demling 1999). However, the non-randomly selected
showed enhanced deposition of leg muscle, but not and non-equivalent control group (n 16, with less
whole body protein, when evaluated in 26 children at severe burns) and the lack of blinding limit the inter-
six months after admission (Eyben et al. 2005). In pretation of this study. Whether improved skin
another study, 35 burned children demonstrated healing at the donor site will lead to improved overall
increased synthesis of constitutive circulating proteins recovery and specifically promote the healing of
and reduced acute phase reactants as well as a reduced severely burned skin remains unproven. These indef-
requirement for albumin supplementation (Thomas inite findings need to be considered in the light of
et al. 2004). However, neither study demonstrated experimental evidence suggesting that androgens may
improved clinical outcomes. retard skin wound healing (Gilliver et al. 2008; 2009).
Other studies, however, have been unable to detect Various studies have also examined whether
any clinical benefits. One well-designed study ran- androgens may improve recovery from burns injury
domized 48 patients requiring hyperalimentation to in the context that numerous new pharmacological

385
Chapter 17: Androgen therapy in non-gonadal disease

approaches are undergoing evaluation (Gauglitz et al. 17.9 Immune disease


2011). Burns injury is a salient test for the application
Androgen therapy for HIV/AIDS has been investi-
of pharmacological androgen therapy where the
gated mainly for its effects on disease-associated mor-
intent to modify the underlying natural history of
bidity (weight loss, weakness, QoL) rather than to
the disorder is particularly plausible. Recovery from
influence the underlying disease natural history.
burns depends primarily on the extent and severity of
Indeed, randomized, placebo-controlled studies have
the burns injury but also on the impact of the injury
consistently reported no androgen effect on CD4
on vital organ systems either directly (lungs) or indir-
count or viral load (Coodley and Coodley 1997;
ectly (kidneys) during recovery. The gravity of the
Grinspoon et al. 1998; Bhasin et al. 1998; Strawford
underlying disorder may be sufficient to justify the
et al. 1999; Sattler et al. 1999; Rabkin et al. 1999; Dobs
overriding relative contraindications of hepatotoxicity
et al. 1999; Bhasin et al. 2001), with two exceptions
(alkylated androgens) and inappropriate virilization
(Berger et al. 1996; Grinspoon et al. 2000), neither of
(use for women and children). The hypercatabolic
which showed a consistent decrease in both CD4
state present during recovery provides the opportun-
count and viral load. One rationale for androgen
ity for exogenous androgens to ameliorate the clinical
therapy stems from the observation that body weight
course and enhance rehabilitation from the burns
loss is an important terminal determinant of survival
injury. Four placebo-controlled studies have shown
in AIDS and other fatal diseases (Grunfeld and Fein-
consistent clinical and biochemical benefits of oxan-
gold 1992). It has been estimated that death occurs
drolone relative to placebo in burned adults (Wolf
when lean body mass reaches 66% of ideal (Kotler
et al. 2006; Przkora et al. 2007) and children (Murphy
et al. 1989), leading to the proposition that, if andro-
et al. 2004; Jeschke et al. 2007), despite the expected
gens (or other agents including megestrol or GH)
hepatotoxicity of oxandrolone. Large-scale, random-
increased appetite and/or body weight, death may be
ized, well-controlled trials are required to evaluate the
delayed. Given this hypothesis, the effect of androgen
definitive risks and benefits (Miller and Btaiche 2009);
therapy may differ between men with AIDS wasting,
whether the same benefits could be obtained with non-
and those without weight loss.
hepatotoxic androgens also remains to be determined.

17.9.1 AIDS/HIV wasting


17.8.3 Rehabilitation A number of randomized, placebo-controlled studies
Two studies have examined whether short-term of androgen therapy in HIV-positive men with AIDS
pharmacological androgen therapy can improve wasting have reported increased lean mass, but min-
rehabilitation in older men. The first randomized 25 imal effects on total body weight, possibly due to
men scheduled for knee replacement to receive weekly concomitantly reduced fat mass. In the most compre-
doses of 300 mg testosterone enanthate or matched hensive study (Grinspoon et al. 1998; 2000), 51 men
placebo during three weeks before surgery (Amory selected for both weight loss and low serum testo-
et al. 2002). The second randomized 15 men admitted sterone concentration were randomized to receive
to hospital for general physical rehabilitation to receive testosterone enanthate 300 mg or oil-based placebo
weekly injections of 100 mg testosterone enanthate or intramuscularly every three weeks for six months.
placebo for two months (Bakhshi et al. 2000). Small Although total weight, fat mass (DXA), total body
improvements in Functional Independence Measure water content (bioimpedance) and physical function
(FIM) score and strength (hand-grip dynamometry) were not changed by testosterone therapy, fat-free
were reported only in the latter study which, however, mass (DXA), lean mass (total body potassium) and
suffered from the limitations of small sample size, non- muscle mass (urinary creatinine excretion) were all
matching saline placebo and unbalanced groups despite increased (Grinspoon et al. 1998). The increased lean
randomization (Honkanen and Lesser 2001). Further body mass was sustained during the open-label, six
well-controlled studies focusing on rehabilitation month extension (Grinspoon et al. 1999). In contrast,
following elective surgery in older men would be of the other four studies have examined body compos-
interest; however, adverse outcomes of androgen ther- itional changes less comprehensively (Dobs et al.
apy (on ventilator-dependent surgical patients) have 1999; Batterham and Garsia 2001) or not at all
also been reported (Bulger et al. 2004). (Berger et al. 1996; Coodley and Coodley 1997). The

386
Chapter 17: Androgen therapy in non-gonadal disease

first randomized 63 HIV seropositive men suffering times each week throughout the study. In one study
from wasting and weakness to receive either 15 mg or (Bhasin et al. 1998), 61 men were randomized to
5 mg oxandrolone daily or placebo for 16 weeks receive testosterone enanthate 100 mg/wk and/or
(Berger et al. 1996). Both oxandrolone (but not con- resistance exercise for 16 weeks. Among the 49 evalu-
trol) groups demonstrated transient weight gain able men, testosterone or resistance exercise increased
within the first month, peaking at the first week. body weight, thigh muscle volume (MRI), muscle
Subsequently, while the high-dose group maintained strength and lean body mass (deuterium oxide dilu-
mean weight gain and the other groups less so, tion and DXA) compared with the control (placebo,
the within-group variance increased, suggesting no exercise) group, but the combination did not pro-
major within-group heterogeneity in time-course. mote further gains. Quality of life was not altered. In
There was also no clear dose-response relationship. the other study (Grinspoon et al. 2000; Fairfield et al.
A second placebo-controlled crossover study random- 2001), 50 men were randomized to receive testoster-
ized 39 men with HIV-associated weight loss to one enanthate 200 mg/wk and/or resistance exercise
receive injections of either testosterone cypionate for 12 weeks. Among the 43 evaluable men, testoster-
200 mg or placebo (of unstated type) every fortnight one or resistance exercise increased body weight, lean
for three months before crossing over to the other mass (DXA) and some components of strength, and
treatment (Coodley and Coodley 1997). Although reduced fat mass (DXA) (Grinspoon et al. 2000).
testosterone improved one of five aspects of QoL The effect of the combination over testosterone ther-
(overall well-being), there were no changes in the apy or exercise alone was not reported. Another study
other components or in weight. However, the null of 24 men with HIV-associated weight loss treated all
effect could have been due to the lack of washout with progressive resistance exercise and testosterone
between treatments. A third study selected men with enanthate 100 mg each week to suppress endogenous
HIV-associated weight loss with serum testosterone testosterone production and then randomized half to
concentrations in the low-normal range (Dobs et al. additionally receive oxandrolone 20 mg each day or
1999). This multicenter, placebo-controlled study ran- placebo tablets for eight weeks (Strawford et al. 1999).
domized 133 men to receive trans-scrotal testosterone The addition of oxandrolone was reported to increase
patch (delivering nominal 6 mg testosterone per day) lean tissue accrual and strength; however, the lack of a
or matching placebo daily for 12 weeks. Testosterone no-treatment control and the concurrent use of two
treatment did not alter weight or lean mass (bioimped- androgens makes interpretation difficult.
ance); however, inconsistent improvements in QoL Further studies have used a three-arm study
were observed. These findings are supported by a design to examine the influence of aromatization on
study that randomized 15 men to receive nandrolone beneficial androgen effects (Crawford et al. 2003)
decanoate (100 mg/fortnight), megestrol acetate (400 by contrasting the effects of the non-aromatizable
mg/day) or dietary advice alone and reported that pure androgen nandrolone with the aromatizable
nandrolone did not increase weight or lean mass androgen testosterone and placebo (Gold et al. 2006;
(bioimpedance) (Batterham and Garsia 2001). Sardar et al. 2010), with both demonstrating superior
Confirmatory findings were also reported in a well- efficacy of nandrolone. Although consistent with the
designed, multi-center study of 38 HIV-positive proposition that aromatization is unimportant for
women with AIDS wasting (Mulligan et al. 2007). such androgen effects, this conclusion hinges on the
A further study of oxandrolone administration con- difficulty of proving dose equivalence of the two
firms the benefits of an androgen (Stenner et al. androgen regimens.
1998), although there is little justification for using a
hepatotoxic androgen when safer non-alkylated
androgens are equally effective. 17.9.2 HIV without wasting
Studies in men with AIDS wasting have confirmed In HIV-positive men without wasting, androgen-
the additive effect of exercise. Two studies examined induced changes in body composition are more
the effect of im testosterone therapy with or without modest. One study of 41 HIV-positive men selected
exercise. In both, men were selected on the basis of participants for low-normal serum testosterone con-
HIV-associated weight loss and exposed to exercise centrations (but not weight loss) and randomized
consisting of a progressive resistance program three them to 12 months of daily transdermal treatment

387
Chapter 17: Androgen therapy in non-gonadal disease

with testosterone (delivering 5 mg testosterone daily) clinical studies have shown unequivocal benefits of
or placebo patch (Bhasin et al. 1998). Testosterone androgen therapy. The recent availability of recom-
produced a greater reduction in fat mass (DXA) binant human G-CSF/GM-CSF, with its greater
but no difference in lean mass, physical function efficacy and better tolerability reduces the benefits
(strength) or QoL. The additive effect of testosterone from androgen-induced prevention of neutropenia
with exercise has also been examined in HIV-positive to cost-effective, second-line status.
men without weight loss. In this study, all 30 men Androgen therapy appears to have morbidity
with stable weight were treated with supraphysiologi- benefits in some but not all studies. One open,
cal weekly doses of im nandrolone decanoate (200 mg controlled study randomized 33 patients with lung
for the first dose, 400 mg for the second dose and 600 or other non-hormone responsive solid cancers to
mg for all subsequent doses) and randomized, half to standard chemotherapy plus nandrolone decanoate
additionally receive progressive resistance exercise (200 mg weekly) or no additional treatment. In this
three times each week, or not, for 12 weeks (Sattler study androgen therapy produced better maintenance
et al. 1999). Although resistance exercise augmented of body weight, hemoglobin and less transfusion
gains in muscular strength and lean body mass (DXA requirement, but no improved survival or physical
and bioimpedance), there was no additional effect on performance (Spiers et al. 1981). Similarly, a cohort
body weight. The lack of a no-treatment control and of 23 patients with inoperable lung cancer requiring
the unblinded exercise intervention limit the inter- palliative chest radiotherapy were randomized to
pretation of this study. In this population of men with receive or not to receive additional treatment with
relatively stable HIV infection, another study exam- nandrolone phenylpropionate (loading dose 100 mg
ined the acceptability of testosterone route of delivery followed by 50 mg weekly during hospitalization).
among 30 HIV-positive men treated for eight weeks During radiotherapy (4500 cGy), androgen therapy
with im injections every one to two weeks, then maintained higher hemoglobin and lower transfusion
switched to eight weeks of daily application of a requirements (Evans and Elias 1972). In contrast, two
transdermal gel (Scott et al. 2007). The transdermal other studies have failed to demonstrate a definite
route demonstrated more stable blood testosterone benefit of androgen therapy. In one study of 40 adults
levels and was preferred, although the one-way switch (including 9 women) who had undergone esophageal
design was a limitation. resection for carcinoma, subjects were randomized to
A well-designed study, examining 75 HIV-positive, receive im injections of nandrolone decanoate 50 mg
non-wasted men with abdominal obesity treated with or oil-based placebo every three weeks for three
10 g transdermal testosterone gel daily for 24 weeks, months commencing one month after resection
observed reduction in total body and subcutaneous fat (Darnton et al. 1999). No treatment effect in weight,
depots but lesser effects on visceral fat mass (Bhasin appetite or mid-arm circumference was detected,
et al. 2007). although appetite improved in both groups with time.
In the other study, 37 patients with unresectable non-
small cell lung cancer requiring standard combina-
17.10 Malignant disease tion chemotherapy were randomized to receive, or
Androgen therapy could influence mortality from not, additional treatment with nandrolone decanoate
malignant disease via direct antitumor effects, or (200 mg weekly for four weeks). Androgen therapy
improve morbidity by maintaining weight, hemo- was associated with only a non-significant statistical
globin, neutrophil count, muscle mass and bone trend towards improved survival (median 8.2 vs. 5.5
mass through its known actions. Reduced morbidity months) and less weight loss, but no improvement
may also augment treatment by creating greater tol- in marrow function (Chlebowski et al. 1986). The
erance for more aggressive cytotoxic therapy. Despite subtherapeutic dose employed by the first study
encouraging results from animal models and uncon- (Darnton et al. 1999) and the greater myelosuppres-
trolled clinical reports, human studies are unconvin- sion resulting from aggressive modern combination
cing. Although older studies demonstrate a consistent chemotherapy in the second study (Chlebowski et al.
but modest effect of androgen therapy in reducing 1986) may have negated any morbidity benefits.
the magnitude, duration and/or complications from Another study has examined the effect of andro-
chemotherapy-induced neutropenia, few well-controlled gen, progestin or corticosteroid treatment for an

388
Chapter 17: Androgen therapy in non-gonadal disease

indefinite period of time on appetite and weight in chemotherapy alone or in combination with BCG vac-
475 men and women with weight loss due to cination, stanozolol (0.1 mg/kgd) or BCG vaccination
advanced incurable cancer (Loprinzi et al. 1999). Sub- plus stanozolol. After three years of follow-up, all four
jects were stratified by cancer type, prognosis and arms had similar rates of remission and adverse events
degree of weight loss before being randomized to (Mandelli et al. 1981).
receive fluoxymesterone 20 mg/day or megestrol acet- Androgen therapy continues to have an estab-
ate 800 mg/day or dexamethasone 3 mg/day in a lished role in late-stage advanced breast cancer, usu-
double-blind fashion for a median duration of two ally as a late option after failure of other hormonal
months. Although survival or QoL was equivalent therapies and when the virilizing side-effects may be
between groups, fluoxymesterone at the dose admin- more acceptable. Few recent studies have focused on
istered was significantly inferior for appetite stimula- androgen therapy, so that clinically it now occupies
tion and tended to result in less weight gain. a residual but diminishing role relative to modern
Furthermore, hirsutism and virilization were major hormonal and cytotoxic chemotherapy for breast
problems occurring in about 10% of all women. The cancer. The frequency and prognostic significance of
role of fluoxymesterone to stimulate appetite is AR expression in breast cancer (Gucalp and Traina
doubtful given the clear superiority of other agents 2010), together with recent experimental advances in
and the dubious QoL consequences of this indication. understanding the role of androgen action in female
A study of 35 men with hematological malignan- reproductive physiology (Walters et al. 2010), may
cies in complete remission following treatment with lead to re-examining the role of androgens in female
cytotoxic chemotherapy evaluated the role of andro- hormone-dependent cancers.
gen therapy for compensated Leydig cell failure. Men
with low-normal circulating testosterone and raised
LH concentrations were randomized (single blind) to 17.11 Respiratory disease
receive, for 12 months, transdermal placebo or testos-
terone patches (2.55 mg/day), dose titrated to main- 17.11.1 Chronic obstructive lung disease
tain a serum testosterone concentration of >20 nM Advanced chronic airflow limitation is associated
(Howell et al. 2001). Testosterone treatment did not with weight loss and muscle depletion, possibly
alter bone turnover markers, hip, spine or forearm due to the increased energy required for breathing,
BMD (quantitative computed tomography and DXA), or reduced serum testosterone concentrations
lean mass or fat mass (DXA), mood (hospital anxiety (Kamischke et al. 1998). Interventions aimed at
and depression scale) or sexual function. However, improving muscle bulk such as nutrition, exercise or
two out of five components of the multidimensional androgens may therefore have an impact on the mor-
fatigue inventory were improved (activity was bidity and/or mortality of the underlying respiratory
increased and physical fatigue was reduced). These disease. A few well-controlled studies examining the
inconsistent and minor effects were supported by a effects of androgens have been reviewed (Svartberg
case control study showing minimal differences based 2010). Six placebo-controlled, parallel-group studies
on lower serum testosterone concentrations in similar (Schols et al. 1995; Ferreira et al. 1998; Creutzberg
men (Howell et al. 2000), suggesting that androgen et al. 2003; Svartberg et al. 2004; Casaburi et al. 2004;
replacement therapy offers little objective benefit Sharma et al. 2008) have been conducted involving
for men with compensated Leydig cell failure post- 395 patients treated for 827 weeks with injectable
cytotoxic therapy. nandrolone decanoate (3, 50 mg every fortnight),
Pharmacological androgen therapy has also been testosterone enanthate (2, 250 or 400 mg per month)
evaluated for maintenance therapy for acute non- or oral stanozolol (1, 12 mg daily). None showed any
lymphocytic leukemia (ANLL), on the basis that improvement in objective lung function, although
enhanced proliferation of residual normal hematopoie- five of six showed improvement in muscle. As in
tic precursors would competitively suppress the growth other medical conditions, it is possible that the effects
of the leukemic clones. Among 114 / 212 patients with of androgen may be due to mood elevation and
newly diagnosed ANLL who obtained complete remis- improved coping mentality and behaviors rather
sion after standard induction chemotherapy, 82 agreed than any direct impact on the natural history of the
to be randomized to undergo standard maintenance underlying disease.

389
Chapter 17: Androgen therapy in non-gonadal disease

The largest well-conducted prospective study dem- using positive air pressure to maintain patency
onstrated that short-term, low-dose androgen therapy (Grunstein et al. 1989). A case report has indicated
(nandrolone decanoate) augmented the effects of nutri- that sleep apnea may be precipitated in an obese man
tional supplementation in patients with moderate to by testosterone administration (Sandblom et al.
severe chronic airways disease (Schols et al. 1995). 1983), and high-dose testosterone can adversely affect
From 233 consecutive patients with stable, moderate sleep patterns in older men (Liu et al. 2003b). These
to severe and bronchodilator-unresponsive pulmonary considerations indicate caution is required when
disease admitted to an intensive pulmonary rehabili- considering pharmacological androgen therapy, espe-
tation program, 217 were randomized into three cially in obese men who are at higher risk of undiag-
groups. These were to receive eight weeks of treatment nosed obstructive sleep apnea. Nevertheless, the
with (1) placebo injections; (2) a nutritional supple- actual risk of precipitating sleep apnea by testosterone
ment (one high-fat, high-calorie drink daily) plus administration to unselected older men remains low.
placebo injections; or (3) a nutritional supplement plus Enquiring about heavy snoring and irregular sleep
androgen injections (nandrolone decanoate (50 mg breathing and daytime somnolence are usually suit-
men, 25 mg women)) with im injections given fort- able screening tests.
nightly. Participants were also stratified according to Androgen therapy has been reported to increase
the degree of baseline muscle depletion (body weight sleep arousals from disordered breathing in a ran-
< 90% and/or lean mass < 67% ideal, or not) at entry. domized crossover study of 11 hypogonadal men
During the study all patients underwent a standardized receiving testosterone enanthate (200400 mg per
exercise program. Both nutrition and androgen ther- fortnight) or no therapy. This study compared som-
apy increased body weight over placebo, with andro- nography during androgen therapy (37 days after a
gen therapy having more prominent effects on lean testosterone injection) with a no-treatment group
body mass and respiratory muscle strength, although consisting of patients after withdrawal (mean 53 days
there was no measurable improvement in submaxi- post-injection) of androgen therapy (Schneider et al.
mal exercise tolerance nor any major adverse effects. 1986). Anatomical and functional evaluation of the
The lack of an androgen-alone arm and blinding with upper-airway patency in four patients showed no
respect to nutritional supplementation made it diffi- treatment-related difference, but this finding is incon-
cult to evaluate the impact of androgen therapy rela- clusive due to the small sample size. Another retro-
tive to improved nutrition. After four years, follow-up spective study examined the prevalence of obstructive
of 203 of these men revealed no treatment effect on sleep apnea in hemodialyzed men and the potential
survival (Schols et al. 1998); however in a post-hoc role of testosterone ester injections in its causation
analysis, those with larger increases in weight (includ- (Millman et al. 1985). Obstructive sleep apnea symp-
ing 24% of the initial placebo group) had a signifi- toms were common (12 / 29, 41%), particularly in those
cantly decreased mortality risk. receiving regular testosterone enanthate injections
Improvement in underlying pulmonary disease (250 mg weekly) to stimulate erythropoiesis (9/12,
itself may ameliorate the gonadal dysfunction of sys- 75%) compared with those not receiving testosterone
temic disease. In one study of men with chronic (6/17, 35%). Withdrawal of testosterone, however, did
obstructive pulmonary disease with severe hypoxia not alter the signs or symptoms of sleep apnea in the
and impotence, long-term oxygen therapy improved five men studied both during and two months after
total and free testosterone and lowered SHBG (without cessation of testosterone treatment. This suggests that
changes in LH or FSH) in five men who had improved testosterone ester injections may not be a regular
sexual function. The remaining seven who had unim- precipitant of obstructive sleep apnea. This is corrob-
proved sexual function had no changes in circulating orated by further surveillance showing that sleep
hormone concentrations (Aasebo et al. 1993). apnea is common among patients with chronic renal
failure even before commencement of dialysis or tes-
tosterone treatment (Kimmel et al. 1989).
17.11.2 Obstructive sleep apnea The effect of androgen therapy on sleep and
Obstructive sleep apnea is associated with reduced breathing has been examined in only three random-
blood testosterone (Liu et al. 2007); a feature that is ized, placebo-controlled studies. In the largest study,
rectified by mechanical splinting of the upper airways among 108 older men (Snyder et al. 1999) randomized

390
Chapter 17: Androgen therapy in non-gonadal disease

to receive a dose-titrated testosterone patch (approxi- status, but the lack of control group or fixed regimen
mately 6 mg/day) or matching placebo for three years, are major limitations (Wulfsohn et al. 1964). A small,
sleep breathing did not deteriorate, although the double-blind study of 15 steroid-dependent asthmatic
tracking device may lack sensitivity (Portier et al. boys randomized to ethylestrenol (0.1 mg/kgd) or
2000), and sleep architecture was not examined. In placebo for 12 months reported a significant improve-
a small, randomized, placebo-controlled study, 10 ment in peak expiratory flow rate in the androgen
men rendered acutely hypogonadal with leuprorelin group compared with the placebo group (Kerrebijn
(Leibenluft et al. 1997) were randomized to receive and Delver 1969). Despite the claim of no acceleration
testosterone enanthate 200 mg every fortnight or oil of bone maturation (according to the ratio of
placebo for four weeks. Testosterone did not alter bone-age/height velocity) in this older study, the
overnight plethysmography-determined sleep par- safety of such androgen therapy in boys prior to
ameters (except that time slept in stage 4 sleep was completion of puberty is very doubtful, and androgen
lengthened), but the effects on breathing were not therapy has no place in the modern treatment of
reported. Whether the frequent overnight blood adolescent asthma.
sampling may have influenced sleep is not clear.
In the only randomized, placebo-controlled study
to examine both sleep and breathing, 17 commu- 17.12 Neurological disease
nity-dwelling healthy men over the age of 60 were
randomized to receive three injections of im testo- 17.12.1 Cognitive function
sterone esters at weekly intervals (500 mg, 250 mg Studies over the last decade provided promising evi-
and 250 mg) or matching oil-based placebo, and dence for beneficial effects of testosterone on cogni-
then crossed over to the other treatment after eight tive function, with potential to ameliorate its decline
weeks washout (Liu et al. 2003b). Testosterone treat- with aging (Cherrier et al. 2001; 2003; 2004; 2005a).
ment shortened sleep ( 1 hour), worsened sleep Furthermore, two pilot studies have been performed:
apnea (by 7 events/hour) and increased the dur- one in 15 men with Alzheimers disease and 17 with
ation of hypoxemia ( 5 min/night), but did not mild cognitive impairment (Cherrier et al. 2005b),
worsen function (driving ability and psychomotor and another in 16 men with Alzheimers disease and
performance). Together these studies suggest that 22 healthy controls (Lu et al. 2006), both showing
high-dose administration of testosterone esters may modest, selective improvement in some but not all
have adverse effects on sleep and breathing in the cognitive domains. However, subsequent studies in
short-term; however, the effects of longer-term use otherwise healthy older men have failed to replicate
of lower, more physiological testosterone doses the Cherrier studies (Kenny et al. 2004; Haren et al.
remains unknown. 2005; Gray et al. 2005; Vaughan et al. 2007), with one
A low frequency of obstructive sleep apnea com- study showing detrimental effects of testosterone
plicating androgen therapy as an idiosyncratic effect (Maki et al. 2007) on cognitive function. One limita-
cannot be excluded. Whether this idiosyncratic reac- tion of the original positive studies was their lack of
tion is related to the pharmacokinetics of the testos- proper masking for oil-based injectable testosterone
terone formulation used, such as the extreme peak by using a saline placebo, which is a serious limitation
serum testosterone following im injections, has yet to for studies with subjective, psychological or effort-
be determined. Whether similar effects would occur dependent end-points (Handelsman 2010b). More-
with more physiological testosterone formulations over, large observational (LeBlanc et al. 2010) and
remains to be established in properly controlled clin- interventional (Young et al. 2010) studies found
ical trials, although the low frequency of such reac- no corroborative effect of sex steroids on cognitive
tions would require very large studies. function at any age. Testosterone may have socio-
behavioral effects (Zak et al. 2009), and it remains
possible that non-specific mood or other psycho-
17.11.3 Asthma logical consequences of androgen deprivation may
Few studies have examined androgen effects in disrupt cognitive function. More powerful and specif-
asthma. One uncontrolled study of asthmatic women ically designed studies may be informative (Warren
administered testosterone reported improved clinical et al. 2008; Cherrier 2009).

391
Chapter 17: Androgen therapy in non-gonadal disease

17.12.2 Headache placebo for six weeks (Itil et al. 1984). Both groups
improved equally in scores for global clinical impres-
The role of androgen withdrawal and therapy in men
sion, physicians checklist for depression, self-rating
with cluster headache, an almost exclusively male
and Hamilton depression rating, and there were no
disorder, has been examined in two controlled stud-
differences in objective measures (EEG, plasma
ies. In one, 60 men with chronic cluster headache
monoamine oxidase levels).
were randomized single-blind to treatment with a
A systematic review and meta-analysis has evalu-
single dose of a GnRH analog (3.75 mg leuprorelin
ated 23 studies using an androgen for depression
depot) or vehicle injection (Nicolodi et al. 1993a).
(Zarrouf et al. 2009). In addition to 18 studies using
Self-reported frequency, intensity and duration of
testosterone, other studies utilized mesterolone (2),
headache as well as sexual activity declined progres-
methyltestosterone (1), and the androgen precursor
sively during three successive 10-day periods after
DHEA (2). After excluding 16 studies not meeting
injection, compared with pre-injection baseline in
study-design quality criteria, 7 studies underwent
those treated with leuprorelin; whereas there was
formal meta-analysis to show a significant benefit of
no change in placebo-treated men. The therapeutic
testosterone relative to placebo. Subgroup analysis
response was delayed in onset, corresponding tem-
showed that significant benefit was confined to the
porally to the onset of castrate testosterone concen-
hypogonadal (vs. eugonadal) subgroup. However, this
trations, and the benefit persisted in most men for the
meta-analysis was significantly flawed by including
one-month post-treatment follow-up period, while
the largest clinical trial which used DHEA, a very
no changes were noted at any stage in the placebo
weak androgen in women but ineffective in men, as
group. As headache is a remitting illness with subject-
well as including women and men (Rabkin et al.
ive study end-points, the unmasking of active drug
2006), whereas the other 6 studies included only
by the regular occurrence of sexual dysfunction in the
men and used effective doses of testosterone. Due to
treated group undermines the validity of the placebo-
the meta-analysis weighting by study size, this study
control group. The surprising absence of a placebo
had strongly disproportionate influence (44.5%
effect in the intended control group reinforces the
weighting vs. others 5.118.1%), and the meta-analysis
possibility of an observer bias. Subsequently, another
conclusions may not be robust to excluding the DHEA
study was conducted in which 12 men with chronic
study. Some additional studies reported since the meta-
cluster headache and 12 non-headache controls
analysis included one study of 23 men with mild
underwent treatment with very high-dose androgen
depression using injectable testosterone (200 mg per
therapy (testosterone propionate 100 mg daily) for
10 days) for six weeks (Seidman et al. 2009); however,
14 days (Nicolodi et al. 1993b). Remarkably, this
some significant improvement attributed to testoster-
produced a dramatic increase in self-reported sexual
one cannot exclude the effects of expectation due to the
activity in the cluster headache group, but not the
use of inadequate masking (saline placebo for oil-based
control group. These curious findings warrant more
testosterone injections) (Handelsman 2010b). More
rigorous study with a double-blind study design util-
conclusively, the largest study so far reported ran-
izing more objective end-points.
domizing 100 depressed men who had responded
poorly to conventional antidepressant therapy to
17.12.3 Depression adjunctive treatment with testosterone or matching
Testosterone has long been considered effective for placebo gel for six weeks. This study observed no
treatment of depression (Altschule and Tillotson significant difference in mood or measures of depres-
1948). Oral mesterolone was the first androgen studied sion (Pope et al. 2010), although there was significant
for antidepressive effects. Laboratory evidence that a improvement in male sexual function (Amiaz et al.
single dose of mesterolone (125 mg) mimics the 2011). Whether subgroups of depressed men or spe-
effects of tricyclic antidepressants on the electroenceph- cific features of their depression (e.g. sexual function)
alogram led to a patent predicting that androgens might may respond more effectively remains to be better
have beneficial effects on clinical depression (Itil et al. defined by further well-controlled clinical trials.
1974). However, this was not confirmed in a double- The relatively non-specific mood-elevating effects
blind clinical trial which randomized 52 depressed men of testosterone may be the basis for findings such
to treatment with mesterolone (150450 mg daily) or as a significant improvement relative to placebo in

392
Chapter 17: Androgen therapy in non-gonadal disease

immediate post-operative symptoms following open androgen effects (Pae 2009). On the contrary, a pilot
prostatectomy (Pourmand et al. 2008). study proposed the adjunctive use of estrogen therapy
for men with schizophrenia (Kulkarni et al. 2011)
based on the claim that estradiol treatment has shown
17.12.4 Other neurological disease promise in treatment of women with schizophrenia
Observational studies of men with epilepsy, usually on (Kulkarni et al. 2008). Such pharmacological estrogen
anticonvulsant treatment, demonstrate impairment of therapy, creating an androgen-deficient state, causes
male reproductive function including lowered blood increased adverse cardiovascular effects which neces-
testosterone levels and reduced sexual function and sitated premature termination of studies involving
fertility (Isojarvi 2008), although the effects on other estrogen administration to men (VACURG 1967;
systemic androgen-dependent functions are not well Coronary Drug Project Research Group 1973) and
reported and the role of testosterone is debatable which are also reported with modern medical castra-
(Talbot et al. 2008; Duncan et al. 2009). The principal tion for prostate cancer (Saylor and Smith 2010).
common effect of enzyme-inducing anticonvulsants Larger and longer duration placebo-controlled studies
appears to be manifest as increasing blood SHBG of testosterone administration would be of consider-
levels creating a form of acquired partial androgen able interest.
resistance. However, there have been few studies of Promising preliminary findings from placebo-
testosterone effects in anticonvulsant-treated men controlled pilot studies have been reported for the
with epilepsy (Herzog et al. 1998; 2010). The single use of testosterone in multiple sclerosis (Sicotte et al.
controlled study examined 40 men with epilepsy 2007) and in Parkinsons disease (Okun et al. 2006).
and reduced sexual function and testosterone levels, Further studies would be of great interest.
who all received testosterone (im testosterone cypio-
nate 300 mg fortnightly) for 12 weeks and were then
randomized to receive an added aromatase inhibitor 17.13 Vascular disease
(anastrozole) or matching placebo tablet (Herzog The effect of androgen therapy in coronary artery
et al. 2010). All participants demonstrated significant (Wu and von Eckardstein 2003) and cardiovascular
improvements in mood and QoL measures as well as (Liu et al. 2003a) disease are reviewed in Chapter 10.
in reduced seizure frequency. However, the lack of a This section will review studies of androgen therapy
no-testosterone control group provides little basis for peripheral vascular venous disease.
for valid interpretation of the beforeafter compari- The use of androgen therapy in acute or chronic
sons as indicative of testosterone effects. Despite some venous disease arises from their fibrinolytic effect,
trends, there were no significant effects of aromatase which may reduce venous fibrin plugging. One study
inhibition on any end-point, although the study of chronic venous insufficiency, aiming to test
may have been underpowered for this analysis. Fur- whether androgen therapy would reduce the rate of
ther placebo-controlled studies of testosterone would venous ulceration, involved 60 patients with venous
be of considerable interest to determine whether skin changes but no ulceration being treated with
anticonvulsant treatment does represent an acquired below-knee compression stockings as standard ther-
androgen-resistant state. apy (McMullin et al. 1991). They were randomized to
The potential use of testosterone as adjunct ther- receive either stanozolol (10 mg daily) or placebo
apy for men with schizophrenia has been evaluated tablets for six months, and androgen therapy pro-
by a preliminary study (Ko et al. 2008). This treated duced a significant but modest reduction in the area
30 men with schizophrenia with either transdermal of venous skin changes but no change in prospective
testosterone or placebo gel daily for four weeks rate of new ulcers or skin oxygenation. The side-
and showed improvement in negative schizophrenic effects comprised mostly virilization, presumably
symptoms. An inconclusive Cochrane analysis due to stanozolol treatment of women.
entitled Testosterone for Schizophrenia only Another prospective, two-center study examined
included three studies using DHEA, an ineffective the role of androgen therapy in prevention of post-
androgen precursor, and none with testosterone operative deep venous thrombosis (DVT). In this
(Elias and Kumar 2007). Confusing DHEA with the study 200 patients scheduled for elective major
potent androgen testosterone distorts discussion of abdominal surgery were randomized into three

393
Chapter 17: Androgen therapy in non-gonadal disease

groups (Zawilska et al. 1990). The first received inhaled reviewed in Chapter 11. Studies of testosterone
heparin (800 units/kg) one day prior to surgery alone; a administration in the metabolic syndrome (Corona
second group received the same dose of inhaled heparin et al. 2011a) and type 2 diabetes (Corona et al. 2011b)
plus a single injection of nandrolone phenylpropionate have been subjected to meta-analysis summaries.
(50 mg); and the third group received standard heparin However, although massive obesity is associated with
prophylaxis (5000 units twice daily, subcutaneously). lowering of total testosterone, there are few well-
Treatments were from the day before surgery until the designed, placebo-controlled clinical trials evaluating
fifth post-operative day. Using daily 125I-fibrinogen androgen therapy in men with uncomplicated obesity.
scanning to detect DVT in 183 evaluable patients, there A series of studies by Marin et al. has raised
was no significant difference in post-operative DVT or interesting questions about the role of pharmaco-
clinically significant bleeding episodes among the three logical androgen therapy in obesity. A pilot study
groups. Unfortunately the study had major between- reported reduced waist/hip circumference and
center differences and used a suboptimal detection improved insulin sensitivity following three months
method. It was also underpowered to reliably evaluate transdermal treatment with testosterone (250 mg in
the claim that addition of nandrolone to nebulized 10 g gel daily) in eight men, but not with DHT (250
heparin was as effective as standard heparin but with mg in 10 g gel daily) in nine men (Marin et al. 1992).
much lower bleeding risk. Larger and better-designed The study design, lacking placebo controls or any
studies of the effects of androgen therapy on venous dose finding, did not allow any conclusion as to
disease in men seem warranted. whether this difference arose from differences in skin
bioavailability or androgen type (aromatizable or not)
or potency. The same investigators then reported a
17.14 Body weight double-blind study in which 27 middle-aged men
17.14.1 Wasting with abdominal obesity were randomized to placebo,
Many older studies examined the role of androgen testosterone or DHT treatment by daily topical appli-
therapy to augment body weight in patients with cation of a transdermal gel (125 mg in 5 g gel daily)
wasting or cachexia from a variety of underlying for nine months (Marin et al. 1995). Testosterone
medical diseases, as well as for cosmetic reasons in treatment inhibited lipid uptake into adipose tissue
otherwise healthy people. For example, one double- triglycerides, decreased lipoprotein lipase activity,
blind study treated 28 healthy men and women and reduced visceral fat stores (CT scan) and increased
26 male patients with wasting associated with chronic euglycemic clamp insulin sensitivity compared with
diseases (e.g. tuberculosis, chronic degenerative dis- DHT and placebo groups (Marin 1995). The results of
orders) with placebo or one of two doses (25 mg Marin et al. were not confirmed by another study
or 50 mg daily) of norethandrolone for 12 weeks which randomized 30 obese middle-aged men into
(Watson et al. 1959). The placebo group subsequently three groups to receive oral oxandrolone (10 mg/
also crossed over to active treatment for another 12 day), testosterone enanthate (150 mg) injections fort-
weeks. Compared with placebo, both androgen groups nightly or placebo treatments for nine months using a
had significantly improved body weight gain and double-dummy, double-blinded design (Lovejoy et al.
reported improved appetite and well-being, but there 1995). Due to lowering of HDL-C by oral oxandro-
was no dose-response relationship. Most patients had lone, a monitoring committee required the oxandro-
abnormal bromosulfophthalein retention, and nearly lone arm to be switched to injections of nandrolone
all women experienced some virilization. Very few decanoate (30 mg) fortnightly. None of the androgens
other studies, however, were well-controlled, and the (oxandrolone, nandrolone, testosterone) had any con-
end-point of weight gain has little validity in isolation sistent overall effect on muscle or fat mass, but the
outside the context of the overall objectives of medical interim change in study design reduced its power. The
management for specific illnesses (see HIV/AIDS). discrepancies between these studies require clarifica-
tion with large sample size, longer duration and more
clinically meaningful end-points.
17.14.2 Obesity Human chorionic gonadotropin has been widely
The effects of testosterone on obesity and its associ- used since the 1960s in ad-hoc and unproven low-
ated metabolic syndrome and type 2 diabetes are dose regimens in combination with a low calorie diet

394
Chapter 17: Androgen therapy in non-gonadal disease

to reduce obesity in middle-aged men (Young et al.


1976; Lijesen et al. 1995). A meta-analysis of con- maintaining hemoglobin and reducing
transfusion dependence.
trolled studies (Lijesen et al. 1995) concurs with the
 In anemia of end-stage renal failure, androgen
largest available single study (Young et al. 1976) that
therapy is cheaper than, and augments the
such low-dose hCG therapy is ineffective and has no effects of, EPO, but whether it is equally or less
valid role in the treatment of obesity. effective remains controversial. Restricting the
use of androgen therapy to older men has the
17.15 Key messages most favorable riskbenefit.
 Androgen replacement therapy aims to replicate  Androgen therapy prevents acute episodes of
but not exceed tissue androgen exposure of hereditary angioedema and probably chronic
eugonadal men and hence is limited to urticaria.
testosterone in physiological doses. Although  Many important questions and opportunities
chronic diseases may cause mild androgen remain for androgen therapy in non-gonadal
deficiency as a non-specific consequence of disease, but careful clinical trials are essential for
systemic illness, androgen replacement therapy proper evaluation.
may influence morbidity but is unlikely to  Traditional indications for androgen therapy
improve mortality. (e.g. osteoporosis, anemia, advanced breast
 Pharmacological androgen therapy utilizes cancer) persist until more specific and effective
androgens to maximal efficacy within treatments become available. Nevertheless newer
adequate safety limits without regard to indications, lower cost and/or equivalent efficacy
androgen class or dose. Such treatment is judged may still favor androgen therapy in some
by the efficacy, safety and cost-effectiveness circumstances.
standards of other drugs. Very few studies of  The mood-elevating properties of androgen
pharmacological androgen therapy fulfill the therapy may explain or augment adjuvant effects
requirements of adequate study design of androgen therapy on non-gonadal diseases.
(randomization, placebo control, objective  The best opportunities for future evaluation of
end-points, adequate adjuvant use of androgen therapy in men with non-
power and duration). gonadal disease include steroid-induced
 Pharmacological androgen therapy has not osteoporosis, wasting due to AIDS and cancer, and
reduced mortality or altered the natural history of chronic respiratory, rheumatological and
any non-gonadal disease. neurological diseases. In addition, the role of
 Since 17a-alkylated androgens are hepatotoxic, androgen therapy in recovery and/or rehabilitation
other safer oral and parenteral androgens should after severe catabolic illness such as burns, critical
be preferred where possible. illness or major surgery is promising but requires
 Androgen therapy does not improve mortality or more detailed evaluation.
morbidity from acute or chronic alcoholic liver  Future studies of adjuvant androgen therapy
disease. The effects in non-alcoholic liver disease require high-quality clinical data involving
have not been studied. randomization and placebo controls as well as
 Androgen therapy does not improve survival in optimal dose-finding and real, rather than
aplastic anemia but improves morbidity by surrogate, end-points.

17.16 References (1983) Danazol for the treatment of


idiopathic thrombocytopenic
hypoandrogenemia in healthy,
normal weight men. J Clin
Aasebo U, Gyltnes A, Bremnes RM, purpura. N Engl J Med Endocrinol Metab 82:15431548
Aakvaag A, Slordal L (1993) Reversal 308:13961399
of sexual impotence in male patients Altschule MD, Tillotson KJ (1948) The
with chronic obstructive pulmonary Aloi JA, Bergendahl M, Iranmanesh A, use of testosterone in the treatment
disease and hypoxemia with long Veldhuis JD (1997) Pulsatile of depressions. N Engl J Med
term oxygen therapy. J Steroid intravenous gonadotropin-releasing 239:10361038
Biochem Mol Biol 46:799803 hormone administration averts Ambriz R, Pizzuto J, Morales M,
Ahn YS, Harrington WJ, Simon SR, fasting-induced Chavez G, Guillen C, Aviles A
Mylvaganam R, Pall LM, So AG hypogonadotropism and (1986) Therapeutic effect of danazol

395
Chapter 17: Androgen therapy in non-gonadal disease

on metrorrhagia in patients with testosterone-induced erythrocytosis. Berns JS, Rudnick MR, Cohen RM
idiopathic thrombocyotpenic J Clin Endocrinol Metab (1992) A controlled trial of
purpura (ITP). Nouv Rev Fr 95:47434747 recombinant human erythropoietin
Hematol 28:275279 Bacigalupo A, Chaple M, Hows J, Van and nandrolone decanoate in the
Amiaz R, Pope HG, Mahne T, Kelly JF, Lint MT, McCann S, Milligan D, treatment of anemia in patients on
Brennan BP, Kanayama G, Chessells J, Goldstone AH, chronic hemodialysis. Clin Nephrol
Weiser M, Hudson JI, Seidman SN Ottolander J, vant Veer ET, 37:264267
(2011) Testosterone gel replacement Comotti B, Coser P, Broccia G, Bhasin S, Storer TW, Asbel-Sethi N,
improves sexual function in Bosi A, Locasciulli A, Catalano L, Kilbourne A, Hays R, Sinha-Hikim I,
depressed men taking serotonergic Battista R, Arcese W, Shen R, Arver S, Beall G (1998)
antidepressants: a randomized, Carotenuto M, Marmont AM, Effects of testosterone replacement
placebo-controlled clinical trial. Gordon Smith EC (1993) Treatment with a nongenital, transdermal
J Sex Marital Ther 37:243254 of aplastic anaemia (AA) with system, Androderm, in human
Amory JK, Chansky HA, Chansky KL, antilymphocyte globulin (ALG) immunodeficiency virus-infected
Camuso MR, Hoey CT, and methylprednisolone (MPred) men with low testosterone
Anawalt BD, Matsumoto AM, with or without androgens: a levels. J Clin Endocrinol Metab
Bremner WJ (2002) Preoperative randomized trial from the EBMT 83:31553162
supraphysiological testosterone in SAA working party. Br J Haematol
Bhasin S, Woodhouse L, Casaburi R,
older men undergoing knee 83:145151
Singh AB, Bhasin D, Berman N,
replacement surgery. J Am Geriatr Bakhshi V, Elliott M, Gentili A, Chen X, Yarasheski KE, Magliano L,
Soc 50:16981701 Godschalk M, Mulligan T (2000) Dzekov C, Dzekov J, Bross R,
Anderson FH, Francis RM, Testosterone improves Phillips J, Sinha-Hikim I, Shen R,
Peaston RT, Wastell HJ (1997) rehabilitation outcomes in ill older Storer TW (2001) Testosterone
Androgen supplementation in men. J Am Geriatr Soc 48:550553 dose-response relationships in
eugonadal men with osteoporosis: Ballal SH, Domoto DT, Polack DC, healthy young men. Am J Physiol
effects of six months treatment on Marciulonis P, Martin KJ (1991) Endocrinol Metab 281:
bone formation and resorption. Androgens potentiate the effects of E1172E1181
J Bone Miner Res 12:472478 erythropoietin in the treatment of Bhasin S, Parker RA, Sattler F,
Antus B, Yao Y, Liu S, Song E, Lutz J, anemia of end-stage renal disease. Haubrich R, Alston B, Umbleja T,
Heemann U (2001) Contribution of Am J Kidney Dis 17:2933 Shikuma CM (2007) Effects of
androgens to chronic allograft Banerji A, Sloane DE, Sheffer AL testosterone supplementation on
nephropathy is mediated by (2008) Hereditary angioedema: a whole body and regional fat mass
dihydrotestosterone. Kidney Int current state-of-the-art review, V: and distribution in human
60:19551963 attenuated androgens for the immunodeficiency virus-infected
treatment of hereditary men with abdominal obesity. J Clin
Antus B, Yao Y, Song E, Liu S, Lutz J,
angioedema. Ann Allergy Endocrinol Metab 92:10491057
Heemann U (2002) Opposite effects
of testosterone and estrogens on Asthma Immunol 100 (1 Suppl 2): Blamey SL, Garden OJ, Shenkin A,
chronic allograft nephropathy. S19S22 Carter DC (1984) Modification of
Transpl Int 15:494501 Batterham MJ, Garsia R (2001) postoperative nitrogen balance with
A comparison of megestrol acetate, preoperative anabolic steroid. Clin
Atkinson RA, Srinivas-Shankar U,
nandrolone decanoate and dietary Nutr 2:187192
Roberts SA, Connolly MJ, Adams JE,
Oldham JA, Wu FC, Seynnes OR, counselling for HIV associated Bojesen A, Juul S, Birkebaek N,
Stewart CE, Maganaris CN, weight loss. Int J Androl 24:232240 Gravholt CH (2004) Increased
Narici MV (2010) Effects of Belch JJ, Madhok R, McArdle B, mortality in Klinefelter syndrome.
testosterone on skeletal muscle McLaughlin K, Kluft C, Forbes CD, J Clin Endocrinol Metab
architecture in intermediate-frail Sturrock R (1986) The effect of 89:38303834
and frail elderly men. J Gerontol increasing fibrinolysis in patients Bonkovsky HL, Fiellin DA, Smith GS,
A Biol Sci Med Sci 65:12151219 with rheumatoid arthritis: a double Slaker DP, Simon D, Galambos JT
Bachman E, Feng R, Travison T, Li M, blind study of stanozolol. Q J Med (1991) A randomized, controlled
Olbina G, Ostland V, Ulloor J, 58:1927 trial of treatment of alcoholic
Zhang A, Basaria S, Ganz T, Berger JR, Pall L, Hall CD, Simpson hepatitis with parenteral nutrition
Westerman M, Bhasin S (2010) DM (1996) Oxandrolone in and oxandrolone. I. Short-term
Testosterone suppresses hepcidin in AIDS-wasting myopathy. AIDS effects on liver function. Am
men: a potential mechanism for 10:16571662 J Gastroenterol 86:12001208

396
Chapter 17: Androgen therapy in non-gonadal disease

Booij A, Biewenga-Booij CM, Huber- Casaburi R, Bhasin S, Cosentino L, supplementation: effects on


Bruning O, Cornelis C, Jacobs JW, Porszasz J, Somfay A, Lewis MI, cognition in older men. Neurology
Bijlsma JW (1996) Androgens as Fournier M, Storer TW (2004) 64:290296
adjuvant treatment in Effects of testosterone and Cherrier MM, Matsumoto AM,
postmenopausal female patients resistance training in men with Amory JK, Asthana S, Bremner W,
with rheumatoid arthritis. Ann chronic obstructive pulmonary Peskind ER, Raskind MA, Craft S
Rheum Dis 55:811815 disease. Am J Respir Crit Care Med (2005b) Testosterone improves spatial
Bork K, Bygum A, Hardt J (2008) 170:870878 memory in men with Alzheimer
Benefits and risks of danazol in Champlin RE, Ho WG, Feig SA, disease and mild cognitive
hereditary angioedema: a long-term Winston DJ, Lenarsky C, Gale RP impairment. Neurology 64:20632068
survey of 118 patients. Ann (1985) Do androgens enhance the
Chlebowski RT, Herrold J, Ali I, Oktay
Allergy Asthma Immunol response to antithymocyte globulin
E, Chlebowski JS, Ponce AT, Heber
100:153161 in patients with aplastic anemia?
D, Block JB (1986) Influence of
Branda RF, Amsden TW, Jacob HS A prospective randomized trial.
nandrolone decanoate on weight
(1977) Randomized study of Blood 66:184188
loss in advanced non-small cell lung
nandrolone therapy for anemia due Chan PCK, Wei DCC, Tam SCF, cancer. Cancer 58:183186
to bone marrow failure. Arch Intern Chan FL, Yeung WC, Cheng IKP
Choi JB, Loredo JS, Norman D,
Med 137:6569 (1992) Post-transplant
Mills PJ, Ancoli-Israel S,
Brockenbrough AT, Dittrich MO, erythrocytosis: role of erythropoietin
Ziegler MG, Dimsdale JE (2006)
Page ST, Smith T, Stivelman JC, and male sex hormones. Nephrol
Does obstructive sleep apnea
Bremner WJ (2006) Transdermal Dial Transpl 7:137142
increase hematocrit? Sleep Breath
androgen therapy to augment EPO Cherrier MM (2009) Testosterone 10:155160
in the treatment of anemia of effects on cognition in health and
Cicardi M, Castelli R, Zingale LC,
chronic renal disease. Am J Kidney disease. Front Horm Res 37:150162
Agostoni A (1997) Side effects of
Dis 47:251262 Cherrier MM, Asthana S, Plymate S, long-term prophylaxis with
Brubaker LH, Briere J, Laszlo J, Kraut E, Baker L, Matsumoto AM, attenuated androgens in hereditary
Landaw SA, Peterson P, Goldberg J, Peskind E, Raskind MA, Brodkin K, angioedema: comparison of treated
Donovan P (1982) Treatment of Bremner W, Petrova A, and untreated patients. J Allergy
anemia in myeloproliferative LaTendresse S, Craft S (2001) Clin Immunol 99:194196
disorders: a randomized study of Testosterone supplementation
fluoxymesterone v transfusions improves spatial and verbal Conway AJ, Boylan LM, Howe C,
only. Arch Intern Med memory in healthy older men. Ross G, Handelsman DJ (1988)
142:15331537 Neurology 57:8088 A randomised clinical trial of
testosterone replacement therapy in
Buchwald D, Argyres S, Easterling RE, Cherrier MM, Craft S, Matsumoto AH hypogonadal men. Int J Androl
Oelshlegel FJ Jr, Brewer GJ, (2003) Cognitive changes associated 11:247264
Schoomaker EB, Abbrecht PH, with supplementation of
Williams GW, Weller JM (1977) testosterone or dihydrotestosterone Coodley GO, Coodley MK (1997)
Effect of nandrolone decanoate on in mildly hypogonadal men: a A trial of testosterone therapy for
the anemia of chronic hemodialysis preliminary report. J Androl HIV-associated weight loss. AIDS
patients. Nephron 18:232238 24:568576 11:13471352

Bulger EM, Jurkovich GJ, Farver CL, Cherrier MM, Plymate S, Mohan S, Cooperative Group for the Study of
Klotz P, Maier RV (2004) Asthana S, Matsumoto AM, Aplastic and Refractory Anaemias
Oxandrolone does not improve Bremner W, Peskind E, Raskind M, (1979) Androgen therapy of aplastic
outcome of ventilator dependent Latendresse S, Haley AP, Craft S anaemia: a prospective study of 352
surgical patients. Ann Surg (2004) Relationship between cases. Scand J Haematol 22:343356
240:472478; discussion:478480 testosterone supplementation and Copenhagen Study Group for Liver
Camitta BM, Thomas ED, Nathan DG, insulin-like growth factor-I levels Diseases (1986) Testosterone
Gale RP, Kopecky KJ, Rappeport and cognition in healthy older men. treatment of men with alcoholic
JM, Santos G, Gordon-Smith EC, Psychoneuroendocrinology 29:6582 cirrhosis: a double-blind study.
Storb R (1979) A prospective study Cherrier MM, Matsumoto AM, Hepatology 6:807813
of androgens and bone marrow Amory JK, Ahmed S, Bremner W, Corona G, Monami M, Rastrelli G,
transplantation for treatment of Peskind ER, Raskind MA, Johnson Aversa A, Tishova Y, Saad F,
severe aplastic anemia. Blood M, Craft S (2005a) The role of Lenzi A, Forti G, Mannucci E,
53:504514 aromatization in testosterone Maggi M (2011a) Testosterone and

397
Chapter 17: Androgen therapy in non-gonadal disease

metabolic syndrome: a meta- oxandrolone, after severe burn Elias A, Kumar A (2007) Testosterone
analysis study. J Sex Med 8:272283 injury. Burns 25:215221 for schizophrenia. Cochrane
Demling RH, Orgill DP (2000) The Database Syst Rev 3:CD006197
Corona G, Monami M, Rastrelli G,
Aversa A, Sforza A, Lenzi A, anticatabolic and wound healing El-Sadr A, Sabry AA, Abdel-Rahman M,
Forti G, Mannucci E, Maggi M effects of the testosterone analog El-Barnachawy R, Koraitim M
(2011b) Type 2 diabetes mellitus oxandrolone after severe burn (1990) Treatment of primary
and testosterone: a meta-analysis injury. J Crit Care 15:1217 nocturnal enuresis by oral
study. Int J Androl 34:528540 Diskin CJ (2007) Erythropoietin levels androgen mesterolone. A clinical
and androgens use: what is their and cystometric study. Urology
Coronary Drug Project Research
relationship in the correction of 36:331335
Group (1973) The Coronary Drug
Project. Findings leading to anemia? Arch Intern Med 167:309 Evans JT, Elias EG (1972) The
discontinuation of the 2.5-mg day erythropoietic response to anabolic
Dobs AS, Cofrancesco J, Nolten WE,
estrogen group. JAMA 226:652657 therapy in patients receiving
Danoff A, Anderson R, Hamilton
radiotherapy. J Clin Pharmacol
Craig TJ (2008) Appraisal of danazol CD, Feinberg J, Seekins D,
New Drugs 12:101104
prophylaxis for hereditary Yangco B, Rhame F (1999) The use
angioedema. Allergy Asthma Proc of a transscrotal testosterone Eyben FE, Graugaard C, Vaeth M
29:225231 delivery system in the treatment of (2005) All-cause mortality and
patients with weight loss related to mortality of myocardial infarction
Crawford BA, Liu PY, Kean M, human immunodeficiency virus for 989 legally castrated men. Eur
Bleasel J, Handelsman DJ (2003) infection. Am J Med 107:126132 J Epidemiol 20:863869
Randomised, placebo-controlled
Dontas AS, Papanicolaou NT, Eyman RK, Grossman HJ, Chaney RH,
trial of androgen effects on bone
Papanayiotou P, Malamos BK Call TL (1990) The life expectancy
and muscle in men requiring long-
(1967) Long-term effects of anabolic of profoundly handicapped people
term systemic glucocorticoid
steroids on renal functions in with mental retardation. N Engl
therapy. J Clin Endocrinol Metab
the aged subject. J Gerontol J Med 323:584589
88:31673176
22:268273 Fairfield WP, Treat M, Rosenthal DI,
Creutzberg EC, Wouters EF, Mostert
Drinka PJ, Jochen AL, Cuisinier M, Frontera W, Stanley T, Corcoran C,
R, Pluymers RJ, Schols AM (2003)
Bloom R, Rudman I, Rudman D Costello M, Parlman K, Schoenfeld D,
A role for anabolic steroids in the
(1995) Polycythemia as a Klibanski A, Grinspoon S (2001)
rehabilitation of patients with
complication of testosterone Effects of testosterone and exercise on
COPD? A double-blind, placebo-
replacement therapy in nursing muscle leanness in eugonadal men
controlled, randomized trial. Chest
home men with low with AIDS wasting. J Appl Physiol
124:17331742
testosterone levels. J Am 90:21662171
Cutolo M, Balleari E, Giusti M, Intra E, Geriatr Soc 43:899901 Fenichel G, Pestronk A, Florence J,
Accardo S (1991) Androgen Robison V, Hemelt VM (1997)
replacement therapy in male Drosos AA, van Vliet-Dascalopoulos E,
Andonopoulos AP, Galanopoulou V, A beneficial effect of oxandrolone in
patients with rheumatoid arthritis. the treatment of Duchenne
Arthritis Rheum 34:15 Skopouli FN, Moutsopoulos
HM (1988) Nandrolone decanoate muscular dystrophy: a pilot study.
Daniell HW (2006) Erythropoietin (deca-durabolin) in primary Neurology 48:12251226
resistance during androgen Sjogrens syndrome: a double Fenichel GM, Griggs RC, Kissel J,
deficiency. Arch Intern Med blind study. Clin Exp Rheumatol Kramer TI, Mendell JR, Moxley RT,
166:1923; author reply:19231924 6:5357 Pestronk A, Sheng K, Florence J,
Darnton SJ, Zgainski B, Grenier I, Dubowitz V, Kinali M, Main M, King WM, Pandya S, Robison VD,
Allister K, Hiller L, McManus KG, Mercuri E, Muntoni F (2002) Wang H (2001) A randomized
Steyn RS (1999) The use of an Remission of clinical signs in early efficacy and safety trial of
anabolic steroid (nandrolone Duchenne muscular dystrophy on oxandrolone in the treatment of
decanoate) to improve nutritional intermittent low-dosage Duchenne dystrophy. Neurology
status after esophageal resection prednisolone therapy. Eur J Paediatr 56:10751079
for carcinoma. Dis Esophagus Neurol 6:153159 Fenster LF (1966) The nonefficacy of
12:283288 short-term anabolic steroid therapy
Duncan S, Talbot A, Sheldrick R,
Demling RH (1999) Comparison of the Caswell H (2009) Erectile function, in alcoholic liver disease. Ann Intern
anabolic effects and complications sexual desire, and psychological Med 65:738744
of human growth hormone and well-being in men with epilepsy Ferreira IM, Verreschi IT, Nery LE,
the testosterone analog, Epilepsy Behav 15:351357 Goldstein RS, Zamel N, Brooks D,

398
Chapter 17: Androgen therapy in non-gonadal disease

Jardim JR (1998) The influence of 6 hemodialysis patients. Am J Kidney steroids, gonadotrophins and
months of oral anabolic steroids on Dis 30:495500 prolactin in alcoholic cirrhotic men.
body mass and respiratory muscles Gauglitz GG, Williams FN, Herndon Aliment Pharmacol Ther 2:119128
in undernourished COPD patients. DN, Jeschke MG (2011) Burns: Gold J, Batterham MJ, Rekers H,
Chest 114:1928 where are we standing with Harms MK, Geurts TB, Helmyr
Finfer S, Chittock DR, Su SY, Blair D, propranolol, oxandrolone, PM, Silva de Mendonca J, Falleiros
Foster D, Dhingra V, Bellomo R, recombinant human growth Carvalho LH, Panos G, Pinchera A,
Cook D, Dodek P, Henderson WR, hormone, and the new incretin Aiuti F, Lee C, Horban A, Gatell J,
Hebert PC, Heritier S, Heyland DK, analogs? Curr Opin Clin Nutr Metab Phanuphak P, Prasithsirikul W,
McArthur C, McDonald E, Care 14:176181 Gazzard B, Bloch M, Danner SA
Mitchell I, Myburgh JA, Gelfand JA, Sherins RJ, Alling DW, (2006) Effects of nandrolone
Norton R, Potter J, Robinson BG, Frank MM (1976) Treatment of decanoate compared with placebo
Ronco JJ (2009) Intensive versus hereditary angioedema with or testosterone on HIV-associated
conventional glucose control in danazol: reversal of clinical and wasting. HIV Med 7:146155
critically ill patients. N Engl J Med biochemical abnormalities. N Engl Gordon C, Wallace DJ, Shinada S,
360:12831297 J Med 295:14441448 Kalunian KC, Forbess L,
Franke WW, Berendonk B (1997) Gervasio JM, Dickerson RN, Braunstein GD, Weisman MH
Hormonal doping and Swearingen J, Yates ME, Yuen C, (2008) Testosterone patches in the
androgenization of athletes: a secret Fabian TC, Croce MA, Brown RO management of patients with
program of the German Democratic (2000) Oxandrolone in trauma mild/moderate systemic lupus
Republic government. Clin Chem patients. Pharmacotherapy erythematosus. Rheumatology
43:12621279 20:13281334 (Oxford) 47:334338
French Cooperative Group for the Gilliver SC, Ruckshanthi JP, Gore CJ, Clark SA, Saunders PU (2007)
Study of Aplastic and Refractory Hardman MJ, Nakayama T, Nonhematological mechanisms of
Anaemias (1986) Androgen therapy Ashcroft GS (2008) Sex dimorphism improved sea-level performance
in aplastic anaemia: a comparative in wound healing: the roles of sex after hypoxic exposure. Med Sci
study of high and low-doses and of steroids and macrophage migration Sports Exerc 39:16001609
4 different androgens. Scand inhibitory factor. Endocrinology Gray PB, Singh AB, Woodhouse LJ,
J Haematol 36:346352 149:57475757 Storer TW, Casaburi R, Dzekov J,
Freder W, Valent P (2011) Treatment Gilliver SC, Ruckshanthi JP, Hardman Dzekov C, Sinha-Hikim I, Bhasin S
of refractory or relapsed acquired MJ, Zeef LA, Ashcroft GS (2009) (2005) Dose-dependent effects of
aplastic anemia: review of 5alpha-dihydrotestosterone (DHT) testosterone on sexual function,
established and experimental retards wound closure by inhibiting mood and visuospatial cognition in
approaches. Leuk Lymphoma re-epithelialization. J Pathol older men. J Clin Endocrinol Metab
52:14351445 217:7382 90:38383846
Fust G, Farkas H, Csuka D, Varga L, Gluud C, Bennett P, Dietrichson O, Griggs RC, Kingston W, Herr BE,
Bork K (2011) Long-term efficacy of Johnsen SG, Ranek L, Svendsen LB, Forbes G, Moxley RT (1985) Lack of
danazol treatment in hereditary Juhl E (1981) Short-term parenteral relationship of hypogonadism to
angioedema. Eur J Clin Invest and peroral testosterone muscle wasting in myotonic
41:256262 administration in men with dystrophy. Arch Neurol 42:881885
Gascon A, Belvis JJ, Berisa F, Iglesias E, alcoholic cirrhosis. Scand Griggs RC, Pandya S, Florence JM,
Estopinan V, Teruel JL (1999) J Gastroenterol 16:749755 Brooke MH, Kingston W, Miller JP,
Nandrolone decanoate is a good Gluud C, Dejgard A, Bennett P, Chutkow J, Herr BE, Moxley RT
alternative for the treatment of Svenstrup B (1987) Androgens and (1989) Randomized controlled trial
anemia in elderly male patients oestrogens before and following oral of testosterone in myotonic
on hemodialysis. Geriatr Nephrol testosterone administration in male dystrophy. Neurology 39:219222
Urol 9:6772 patients with and without alcoholic Grinspoon S, Corcoran C, Askari H,
Gaughan WJ, Liss KA, Dunn SR, cirrhosis. Acta Endocrinol (Copenh) Schoenfeld D, Wolf L, Burrows B,
Mangold AM, Buhsmer JP, Michael B, 115:385391 Walsh M, Hayden D, Parlman K,
Burke JF (1997) A 6-month study Gluud C, Bennett P, Svenstrup B, Anderson E, Basgoz N, Klibanski A
of low-dose recombinant human Micic S, Copenhagen Study Group (1998) Effects of androgen
erythropoietin alone and in for Liver Diseases (1988) Effect of administration in men with the
combination with androgens for the oral testosterone treatment on AIDS wasting syndrome.
treatment of anemia in chronic serum concentrations of sex A randomized, double-blind,

399
Chapter 17: Androgen therapy in non-gonadal disease

placebo-controlled trial. Ann Intern Hamilton JB, Mestler GE (1969) Derksen RH (2008) Effect of
Med 129:1826 Mortality and survival: comparison dehydroepiandrosterone
Grinspoon S, Corcoran C, Anderson E, of eunuchs with intact men and administration on fatigue, well-
Hubbard J, Stanley T, Basgoz N, women in a mentally retarded being, and functioning in women
Klibanski A (1999) Sustained population. J Gerontol 24:395411 with primary Sjogren syndrome: a
anabolic effects of long-term Handelsman DJ (1985) Hypothalamic- randomised controlled trial. Ann
androgen administration in men pituitary gonadal dysfunction in Rheum Dis 67:9197
with AIDS wasting. Clin Infect Dis chronic renal failure, dialysis, and Hartkamp A, Geenen R,
28:634636 renal transplantation. Endocr Rev Godaert GL, Bijl M, Bijlsma JW,
6:151182 Derksen RH (2010) Effects of
Grinspoon S, Corcoran C, Parlman K,
Handelsman DJ (2010a) Testicular dehydroepiandrosterone on fatigue
Costello M, Rosenthal D, Anderson E,
dysfunction in systemic diseases. In: and well-being in women with
Stanley T, Schoenfeld D, Burrows B,
Nieschlag E, Behre HM, Nieschlag S quiescent systemic lupus
Hayden D, Basgoz N, Klibanski A
(eds) Andrology. Male Reproductive erythematosus: a randomised
(2000) Effects of testosterone and
Health and Dysfunction, 3rd edn. controlled trial. Ann Rheum Dis
progressive resistance training in
Springer-Verlag, Berlin, pp 339364 69:11441147
eugonadal men with AIDS wasting.
A randomized, controlled trial. Ann Handelsman DJ (2010b) Inadequate Hausmann DF, Nutz V, Rommelsheim
Intern Med 133:348355 masking of testosterone. J Am K, Caspari R, Mosebach KO (1990)
Coll Cardiol 55:2290; author Anabolic steroids in polytrauma
Grunfeld C, Feingold KR (1992)
reply:22902291 patients. Influence on renal nitrogen
Metabolic disturbances and wasting
and amino acid losses: a double-
in the acquired immunodeficiency Handelsman DJ (2011) Commentary: blind study. J Parenter Enter
syndrome. N Engl J Med androgens and anabolic steroids: 14:111114
327:329337 the one-headed Janus.
Endocrinology 152:17521754 Hazelton RA, McCruden AB,
Grunstein RR, Handelsman DJ,
Sturrock RD, Stimson WH (1983)
Lawrence SJ, Blackwell C, Handelsman DJ, Dong Q (1993) Hormonal manipulation of the
Caterson ID, Sullivan CE (1989) Hypothalamo-pituitary gonadal axis immune response in systemic
Hypothalamic dysfunction in sleep in chronic renal failure. Endocrinol lupus erythematosus: a drug
apnea: reversal by nasal continuous Metab Clin North Am 22:145161 trial of an anabolic steroid,
positive airways pressure. J Clin
Handelsman DJ, Liu PY (1998) 19-nortestosterone. Ann Rheum Dis
Endocrinol Metab 68:352358
Androgen therapy in chronic renal 42:155157
Gucalp A, Traina TA (2010) Triple- failure. Baillieres Clin Endocrinol Hendler ED, Goffinet JA, Ross S,
negative breast cancer: role of the Metab 12:485500 Longnecker RE, Bakovic V (1974)
androgen receptor. Cancer J Controlled study of androgen
Handelsman DJ, Mackey MA, Howe C,
16:6265 therapy in anemia of patients on
Turner L, Conway AJ (1997)
Hajjar RR, Kaiser FE, Morley JE (1997) Analysis of testosterone implants maintenance hemodialysis. N Engl
Outcomes of long-term testosterone for androgen replacement therapy. J Med 291:10461051
replacement in older hypogonadal Clin Endocrinol (Oxf) 47:311316 Herzog AG, Klein P, Jacobs AR (1998)
males: a retrospective analysis. Hansell DT, Davies JW, Shenkin A, Testosterone versus testosterone and
J Clin EndocrinolMetab testolactone in treating reproductive
Garden OJ, Burns HJ, Carter DC
82:37933796 (1989) The effects of an anabolic and sexual dysfunction in men with
Hall GM, Larbre JP, Spector TD, Perry steroid and peripherally epilepsy and hypogonadism.
LA, Silva JAD (1996) A randomized administered intravenous nutrition Neurology 50:782784
trial of testosterone therapy in males in the early postoperative period. Herzog AG, Farina EL, Drislane FW,
with rheumatoid arthritis. Br J Parenter Enter 13:349358 Schomer DL, Smithson SD, Fowler
J Rheumatol 35:568573 Haren MT, Wittert GA, Chapman IM, KM, Dworetzky BA, Bromfield EB
Hamdy RC, Moore SW, Whalen KE, Coates P, Morley JE (2005) Effect of (2010) A comparison of anastrozole
Landy C (1998) Nandrolone oral testosterone undecanoate on and testosterone versus placebo and
decanoate for men with visuospatial cognition, mood and testosterone for treatment of sexual
osteoporosis. Am J Ther 5:8995 quality of life in elderly men with dysfunction in men with epilepsy
Hamilton JB (1937) Treatment of low-normal gonadal status. and hypogonadism. Epilepsy Behav
sexual underdevelopment with Maturitas 50:124133 7:264271
synthetic male hormone substance. Hartkamp A, Geenen R, Godaert GL, Honkanen L, Lesser GT (2001)
Endocrinology 21:649654 Bootsma H, Kruize AA, Bijlsma JW, Testosterone use for rehabilitation

400
Chapter 17: Androgen therapy in non-gonadal disease

of older men. J Am Geriatr Soc development of polycythemia in erythropoiesis. Eur J Med Res
49:339340 hypogonadal men treated for up to 2:293298
Horl WH (1999) Is there a role for 21 years with subcutaneous Johansen KL, Mulligan K,
adjuvant therapy in patients being testosterone pellets. Eur J Endocrinol Schambelan M (1999) Anabolic
treated with epoetin? Nephrol Dial 162:385390 effects of nandrolone decanoate
Transpl 14:5060 Ishak KG, Zimmerman HJ (1987) in patients receiving dialysis: a
Hosea SW, Santaella ML, Brown EJ, Hepatotoxic effects of the anabolic- randomized controlled trial. JAMA
Berger M, Katusha K, Frank MM androgenic steroids. Semin Liver Dis 281:12751281
(1980) Long-term therapy of 7:230236 Kaltwasser JP, Dix U, Schalk KP,
hereditary angioedema with Isojarvi J (2008) Disorders of Vogt H (1988) Effect of androgens
danazol. Ann Intern Med reproduction in patients with on the response to antithymocyte
93:809812 epilepsy: antiepileptic drug related globulin in patients with aplastic
Hosseinimehr SJ, Zakaryaee V, mechanisms. Seizure 17:111119 anaemia. Eur J Haematol 40:111118
Froughizadeh M (2006) Oral Itil TM, Cora R, Akpinar S, Herrmann Kamischke A, Kemper DE, Castel MA,
oxymetholone reduces mortality WM, Patterson CJ (1974) Luthke M, Rolf C, Behre HM,
induced by gamma irradiation in Psychotropic action of sex Magnussen H, Nieschlag E (1998)
mice through stimulation of hormones: computerized EEG in Testosterone levels in men with
hematopoietic cells. Mol Cell establishing the immediate CNS chronic obstructive pulmonary
Biochem 287:193199 effects of steroid hormones. Curr disease with or without
Howell SJ, Radford JA, Smets EM, Thera Res 16:11471170 glucocorticoid therapy.
Shalet SM (2000) Fatigue, sexual Eur Respir J 11:4145
Itil TM, Michael ST, Shapiro DM,
function and mood following Itil KZ (1984) The effects of Kassmann K, Rappaport R, Broyer M
treatment for haematological mesterolone, a male sex hormone in (1992) The short-term effect of
malignancy: the impact of mild depressed patients (a double blind testosterone on growth in boys
Leydig cell dysfunction. Br J Cancer controlled study. Methods Find Exp on hemodialysis. Clin Nephrol
82:789793 Clin Pharmacol 6:331337 37:148154
Howell SJ, Radford JA, Adams JE, Jaime-Perez JC, Colunga-Pedraza PR, Kavanagh BP, McCowen KC (2010)
Smets EM, Warburton R, Shalet SM Gomez-Ramirez CD, Gutierrez- Clinical practice. Glycemic control
(2001) Randomized placebo- Aguirre CH, Cantu-Rodriguez OG, in the ICU. N Engl J Med
controlled trial of testosterone Tarin-Arzaga LC, Gomez-Almaguer 363:25402546
replacement in men with mild D (2011) Danazol as first-line
Leydig cell insufficiency following Kennedy BJ, Gilbertsen AS (1957)
therapy for aplastic anemia. Ann Increased erythropoiesis induced by
cytotoxic chemotherapy. Clin Hematol 90:523527
Endocrinol (Oxf) 55:315324 androgenic hormone therapy.
Jayson MI, Holland CD, Keegan A, N Engl J Med 256:719726
Idan A, Griffiths KA, Harwood DT, Illingworth K, Taylor L (1991)
Seibel MJ, Turner L, Conway AJ, Kenny AM, Fabregas G, Song C,
A controlled study of stanozolol in Biskup B, Bellantonio S (2004)
Handelsman DJ (2010) Long-term primary Raynauds phenomenon
effects of dihydrotestosterone Effects of testosterone on behavior,
and systemic sclerosis. Ann Rheum depression, and cognitive function
treatment on prostate growth in Dis 50:4147
healthy, middle-aged men without in older men with mild cognitive
prostate disease: a randomized, Jenkins JS (1998) The voice of the loss. J Gerontol A Biol Sci Med Sci
placebo-controlled trial. Ann Intern castrato. Lancet 351:18771880 59:7578
Med 153:621632 Jeschke MG, Finnerty CC, Suman OE, Kenny AM, Kleppinger A, Annis K,
Inkovaara J, Gothoni G, Halttula R, Kulp G, Mlcak RP, Herndon DN Rathier M, Browner B, Judge JO,
Heikinheimo R, Tokola O (1983) (2007) The effect of oxandrolone on McGee D (2010) Effects of
Calcium, vitamin D and anabolic the endocrinologic, inflammatory, transdermal testosterone on bone
steroid in treatment of aged bones: and hypermetabolic responses and muscle in older men with low
double-blind placebo-controlled during the acute phase postburn. bioavailable testosterone levels,
long-term clinical trial. Age Ageing Ann Surg 246:351360; low bone mass, and physical
12:124130 discussion:360362 frailty. J Am Geriatr Soc
Jockenhvel F, Vogel E, Reinhardt W, 58:11341143
Ip FF, di Pierro I, Brown R,
Cunningham I, Handelsman DJ, Reinwein D (1997) Effects of Kent BD, Mitchell PD, McNicholas
Liu PY (2010) Trough serum various modes of androgen WT (2011) Hypoxemia in patients
testosterone predicts the substitution therapy on with COPD: cause, effects, and

401
Chapter 17: Androgen therapy in non-gonadal disease

disease progression. Int J Chron 19-nortestosterone in the therapy of gonadotropin (HCG) in the
Obstruct Pulmon Dis 6:199208 systemic lupus erythematosus: treatment of obesity by means of the
Kerrebijn KF, Delver A (1969) worsened disease after treatment Simeons therapy: a criteria-based
Ethylestrenol (Orgabolin): effects on with 19-nortestosterone in men and meta-analysis. Br J Clin Pharmacol
asthmatic children during lack of improvement in women. 40:237243
corticosteroid treatment. Scand J Rheumatol 19:547555 Liu PY, Death AK, Handelsman DJ
J Respir Dis 68:7077 Lakshman KM, Basaria S (2009) Safety (2003a) Androgens and
Kimmel PL, Miller G, Mendelson WB and efficacy of testosterone gel in cardiovascular disease. Endocr Rev
(1989) Sleep apnea syndrome in the treatment of male 24:313340
chronic renal disease. Am J Med hypogonadism. Clin Interv Aging
Liu PY, Yee BJ, Wishart SM, Jimenez
86:308314 4:397412
M, Jung DG, Grunstein RR,
Ko YH, Lew YM, Jung SW, Joe SH, Lee Langouche L, van den Berghe G (2006) Handelsman DJ (2003b) The
CH, Jung HG, Lee MS (2008) Short- The dynamic neuroendocrine short-term effects of high dose
term testosterone augmentation in response to critical illness. testosterone on sleep, breathing and
male schizophrenics: a randomized, Endocrinol Metab Clin North Am function in older men. J Clin
double-blind, placebo-controlled 35:777791, ix Endocrinol Metab 88:36053613
trial. J Clin Psychopharmacol LeBlanc ES, Wang PY, Janowsky JS, Liu PY, Caterson ID, Grunstein RR,
28:375383 Neiss MB, Fink HA, Yaffe K, Handelsman DJ (2007) Androgens,
Kochakian CD (ed) (1976) Anabolic- Marshall LM, Lapidus JA, Stefanick obesity, and sleep-disordered
Androgenic Steroids. Springer- ML, Orwoll ES (2010) Association breathing in men. Endocrinol Metab
Verlag, Berlin between sex steroids and cognition Clin North Am 36:349363
in elderly men. Clin Endocrinol
Kopera H (1976) Miscellaneous uses of Loprinzi CL, Kugler JW, Sloan JA,
(Oxf) 72:393403
anabolic steroids. In: Kochakian CD Mailliard JA, Krook JE, Wilwerding
(ed) Anabolic-Androgenic Steroids. Leibenluft E, Schmidt PJ, Turner EH, MB, Rowland KM Jr, Camoriano
Springer-Verlag, Berlin, pp 535625 Danaceau MA, Ashman SB, Wehr JK, Novotny PJ, Christensen BJ
TA, Rubinow DR (1997) Effects of (1999) Randomized comparison of
Kotler DP, Tierney AR, Wang J, leuprolide-induced hypogonadism megestrol acetate versus
Pierson RN (1989) Magnitude of and testosterone replacement on dexamethasone versus
body-cell-mass depletion and the sleep, melatonin, and prolactin fluoxymesterone for the treatment
timing of death from wasting in secretion in men. J Clin Endocrinol of cancer anorexia/cachexia. J Clin
AIDS. Am J Clin Nutr 50:444447 Metab 82:32033207 Oncol 17:32993306
Krauss DJ, Taub HA, Lantinga LJ, Leleu X, Terriou L, Duhamel A, Lovejoy JC, Bray GA, Greeson CS,
Dunsky MH, Kelly CM (1991) Risks Moreau AS, Andrieux J, Dupire S, Klemperer M, Morris J, Partington
of blood volume changes in Coiteux V, Berthon C, Micol JB, C, Tulley R (1995) Oral anabolic
hypogonadal men treated with Guieze R, Facon T, Bauters F (2006) steroid treatment, but not
testosterone enanthate for erectile Long-term outcome in acquired parenteral androgen treatment,
impotence. J Urol 146:15661570 aplastic anemia treated with an decreases abdominal fat in obese,
Krskemper HL (1968) Anabolic intensified dose schedule of horse older men. Int J Obes 19:614624
Steroids. Academic Press, New York antilymphocyte globulin in
combination with androgens. Ann Lu PH, Masterman DA, Mulnard R,
Kulkarni J, de Castella A, Fitzgerald PB, Cotman C, Miller B, Yaffe K,
Gurvich CT, Bailey M, Hematol 85:711716
Reback E, Porter V, Swerdloff R,
Bartholomeusz C, Burger H (2008) Lewis L, Dahn M, Kirkpatrick JR Cummings JL (2006) Effects of
Estrogen in severe mental illness: a (1981) Anabolic steroid testosterone on cognition and mood
potential new treatment approach. administration during nutritional in male patients with mild Alzheimer
Arch Gen Psychiatry 65:955960 support: a therapeutic controversy. disease and healthy elderly men.
Kulkarni J, de Castella A, Headey B, J Parent Ent Nutr 5:6466 Arch Neurol 63:177185
Marston N, Sinclair K, Lee S, Li Bock E, Fulle HH, Heimpel H, Macdonald JH, Marcora SM, Jibani
Gurvich C, Fitzgerald PB, Burger H Pribilla W (1976) Die Wirkung von MM, Kumwenda MJ, Ahmed W,
(2011) Estrogens and men with Mesterolon bei Panmyelopathien Lemmey AB (2007) Nandrolone
schizophrenia: is there a case for und renalen Anaemien. Med Klin decanoate as anabolic therapy in
adjunctive therapy? Schizophr Res 71:539547 chronic kidney disease: a
125:278283 Lijesen GK, Theeuwen I, Assendelft randomized phase II dose-finding
Lahita RG, Cheng CY, Monder C, WJ, Van Der Wal G (1995) The study. Nephron Clin Pract 106:
Bardin CW (1992) Experience with effect of human chorionic c125c135

402
Chapter 17: Androgen therapy in non-gonadal disease

Maddrey WC (1986) Is therapy with stanozolol in the treatment of therapy does not enhance lean tissue
testosterone or anabolic-androgenic venous insufficiency. Austr accrual in men with human
steroids useful in the treatment of N Zealand Surgery 61:306309 immunodeficiency virus-associated
alcoholic liver disease? Hepatology Mendenhall CL, Anderson S, Garcia- weight loss: a randomized, double-
6:10331035 Pont P, Goldberg S, Kiernan T, Seeff blind, placebo-controlled,
Maki PM, Ernst M, London ED, LB, Sorrell M, Tamburro C, multicenter trial. J Clin Endocrinol
Mordecai KL, Perschler P, Durso Weesner R, Zetterman R, Chedid A, Metab 92:563570
SC, Brandt J, Dobs A, Resnick SM Chen T, Rabin L; Veterans Murphy KD, Thomas S, Mlcak RP,
(2007) Intramuscular testosterone Administration Cooperative Study Chinkes DL, Klein GL, Herndon
treatment in elderly men: evidence on Alcoholic Hepatitis (1984) DN (2004) Effects of long-term
of memory decline and altered brain Short-term and long-term survival oxandrolone administration in
function. J Clin Endocrinol Metab in patients with alcoholic hepatitis severely burned children. Surgery
92:41074114 treated with oxandrolone and 136:219224
Maloisel F, Andres E, Zimmer J, Noel prenisolone. N Engl J Med Naik RB, Gibbons AR, Gyde OH,
E, Zamfir A, Koumarianou A, 311:14641470 Harris BR, Robinson BH (1978)
Dufour P (2004) Danazol therapy in Mendenhall CL, Moritz TE, Roselle Androgen trial in renal anaemia.
patients with chronic idiopathic GA, Morgan TR, Nemchausky BA, Proc Eur Dial Transplant Assoc
thrombocytopenic purpura: Tamburro CH, Schiff ER, McClain 15:136143
long-term results. Am J Med CJ, Marsano LS, Allen JI (1993) Najean Y, Pecking A (1979) Refractory
116:590594 A study of oral nutritional support anemia with excess of blast cells:
Mandelli F, Amadori S, Dini E, with oxandrolone in malnourished prognostic factors and effect of
Grignani F, Leoni P, Liso V, patients with alcoholic hepatitis. treatment with androgens or
Martelli M, Neri A, Petti MC, Hepatology 17:564576 cytosine arabinoside. Results of a
Ferrini PR (1981) Randomized Miller JT, Btaiche IF (2009) prospective trial in 58 patients.
clinical trial of immunotherapy and Oxandrolone treatment in adults Cooperative Group for the Study of
androgenotherapy for remission with severe thermal injury. Aplastic and Refractory Anemias.
maintenance in acute non- Pharmacotherapy 29:213226 Cancer 44:19761982
lymphocytic leukemia. Leuk Res Millman RP, Kimmel PL, Shore ET, Najean Y; Joint Group for the Study of
5:447452 Wasserstein AG (1985) Sleep apnea Aplastic and Refractory Anaemias
Marin P (1995) Testosterone and in hemodialysis patients: the lack of (1981) Long-term follow-up in
regional fat distribution. Obes Res 3 testosterone effect on its patients with aplastic anemia.
Suppl 4:609S612S pathogenesis. Nephron 40:407410 A study of 137 androgen-treated
Marin P, Holmang S, Jonsson L, Mooradian AD, Morley JE, Korenman patients surviving more than two
Sjostrom L, Kvist H, Holm G, SG (1987) Biological actions of years. Am J Med 71:543551
Lindstedt G, Bjorntorp P (1992) The androgens. Endocr Rev 8:128 Navarro JF, Mora C (2001) In-depth
effects of testosterone treatment on Muller M, van den Beld AW, van der review effect of androgens on
body composition and metabolism Schouw YT, Grobbee DE, Lamberts anemia and malnutrition in renal
in middle-aged obese men. Int SW (2006) Effects of failure: implications for patients on
J Obes 16:991997 dehydroepiandrosterone and peritoneal dialysis. Perit Dial Int
Marin P, Oden B, Bjorntorp P (1995) atamestane supplementation on 21:1424
Assimilation and mobilization of frailty in elderly men. J Clin Navarro JF, Mora C, Macia M, Garcia J
triglycerides in subcutaneous Endocrinol Metab 91:39883991 (2002) Randomized prospective
abdominal and femoral adipose Muller V, Szabo A, Viklicky O, Gaul I, comparison between erythropoietin
tissue in vivo in men: effects of Portl S, Philipp T, Heemann UW and androgens in CAPD patients.
androgens. J Clin Endocrinol Metab (1999) Sex hormones and gender- Kidney Int 61:15371544
80:239243 related differences: their influence Neff MS, Goldberg J, Slifkin RF, Eiser
Marwaha RK, Bansal D, Trehan A, on chronic renal allograft rejection. AR, Calamia V, Kaplan M, Baez A,
Varma N (2004) Androgens in Kidney Int 55:20112020 Gupta S, Mattoo N (1981)
childhood acquired aplastic Mulligan K, Zackin R, Von Roenn JH, A comparison of androgens for
anaemia in Chandigarh, India. Trop Chesney MA, Egorin MJ, Sattler FR, anemia in patients on hemodialysis.
Doct 34:149152 Benson CA, Liu T, Umbleja T, N Engl J Med 304:871875
McMullin GM, Watkin GT, Coleridge Shriver S, Auchus RJ, Schambelan Neff MS, Goldberg J, Slifkin RF, Eiser
Smith PD, Scurr JH (1991) Efficacy M (2007) Testosterone AR, Calamia V, Kaplan M, Baez A,
of fibrinolytic enhancement with supplementation of megestrol Gupta S, Mattoo N (1985)

403
Chapter 17: Androgen therapy in non-gonadal disease

Anemia in chronic renal Pae CU (2009) Comments on Short- FF (1995) Androgen receptor
failure. Acta Endocrinol Suppl term testosterone augmentation in defects: historical, clinical and
271:8086 male schizophrenics: a randomized, molecular perspectives. Endocr Rev
Nicolodi M, Sicuteri F, Poggioni M double-blind, placebo-controlled 16:271321
(1993a) Hypothalamic modulation trial by Dr Ko and colleagues. Rabkin JG, Wagner GJ, Rabkin R
of nociception and reproduction in J Clin Psychopharmacol 29:194195; (1999) Testosterone therapy for
cluster headache. I. Therapeutic author reply:196197 human immunodeficiency virus-
trials of leuprolide. Cephalgia Palacios A, Campfield LA, McClure positive men with and without
13:253257 RD, Steiner B, Swerdloff RS (1983) hypogonadism. J Clin
Nicolodi M, Sicuteri F, Poggioni M Effect of testosterone enanthate on Psychopharmacol 19:1927
(1993b) Hypothalamic modulation hematopoesis in normal men. Fertil
Rabkin JG, McElhiney MC, Rabkin R,
of nociception and reproduction in Steril 40:100104
McGrath PJ, Ferrando SJ (2006)
cluster headache. II. Testosterone- Parsad D, Pandhi R, Juneja A (2001) Placebo-controlled trial of
induced increase of sexual activity Stanozolol in chronic urticaria: a dehydroepiandrosterone (DHEA)
in males with cluster headache. double blind, placebo controlled for treatment of nonmajor
Cephalgia 13:258260 trial. J Dermatol 28:299302 depression in patients with HIV/
Nieschlag E, Cuppers HJ, Wickings EJ Phillips EH, Ryan S, Ferrari R, Green C AIDS. Am J Psychiatry 163:5966
(1977) Influence of sex, testicular (2003) Estratest and Estratest HS Rambaldi A, Gluud C (2006) Anabolic-
development and liver function on (esterified estrogens and androgenic steroids for alcoholic
the bioavailability of oral methyltestosterone) therapy: a liver disease. Cochrane Database
testosterone. Eur J Clin Invest summary of safety surveillance data, Syst Rev 4:CD003045
7:145147 January 1989 to August 2002. Clin
Ther 25:30273043 Reckelhoff JF, Yanes LL, Iliescu R,
Nieschlag E, Nieschlag S, Behre HM Fortepiani LA, Granger JP (2005)
(1993) Lifespan and testosterone. Pope HG Jr, Amiaz R, Brennan BP, Orr Testosterone supplementation in
Nature 366:215 G, Weiser M, Kelly JF, Kanayama G, aging men and women: possible
Nordmark G, Bengtsson C, Larsson A, Siegel A, Hudson JI, Seidman SN impact on cardiovascular-renal
Karlsson FA, Sturfelt G, Ronnblom (2010) Parallel-group placebo- disease. Am J Physiol Renal Physiol
L (2005) Effects of controlled trial of testosterone gel in 289:F941F948
dehydroepiandrosterone men with major depressive disorder
displaying an incomplete response Reid IR, Wattie DJ, Evans MC,
supplement on health-related Stapleton JP (1996) Testosterone
quality of life in glucocorticoid to standard antidepressant
treatment. J Clin Psychopharmacol therapy in glucocorticoid-treated
treated female patients with men. Arch Intern Med
systemic lupus erythematosus. 30:126134
156:11731177
Autoimmunity 38:531540 Portier F, Portmann A, Czernichow P,
Vascaut L, Devin E, Benhamou D, Reilly JT (2006) Idiopathic
OConnell MD, Tajar A, Roberts SA,
Cuvelier A, Muir JF (2000) myelofibrosis: pathogenesis to
Wu FC (2011) Do androgens play
Evaluation of home versus treatment. Hematol Oncol 24:5663
any role in the physical frailty
of ageing men? Int J Androl laboratory polysomnography in the Rice L (2004) Danazol, idiopathic
34:195211 diagnosis of sleep apnea syndrome. thrombocytopenic purpura, and
Am J Respir Crit Care Med thrombopoietin. Am J Med
Okun MS, Fernandez HH, Rodriguez 162:814818 117:972973; author reply:973
RL, Romrell J, Suelter M, Munson S,
Louis ED, Mulligan T, Foster PS, Pourmand G, Salem S, Karami A, Riedl M, Gower RG, Chrvala CA
Shenal BV, Armaghani SJ, Jacobson Baradaran N, Mehrsai A (2008) (2011) Current medical
C, Wu S, Crucian G (2006) Anabolic-androgenic steroid effects management of hereditary
Testosterone therapy in men with on early morbid symptoms after angioedema: results from a large
Parkinson disease: results of the open prostatectomy: a pilot study. survey of US physicians. Ann
TEST-PD Study. Arch Neurol Aging Male 11:123127 Allergy Asthma Immunol
63:729735 Przkora R, Herndon DN, Suman OE 106:316322.e4
Orr R, Fiatarone Singh M (2004) The (2007) The effects of oxandrolone Sandblom RE, Matsumoto AM, Scoene
anabolic androgenic steroid and exercise on muscle mass and RB, Lee KA, Giblin EC, Bremner
oxandrolone in the treatment of function in children with severe WJ, Pierson DJ (1983) Obstructive
wasting and catabolic disorders: burns. Pediatrics 119:e109e116 sleep apnea induced by testosterone
review of efficacy and safety. Drugs Quigley CA, DeBellis A, Marschke KB, administration. N Engl J Med
64:725750 El-Awady MK, Wilson EM, French 308:508510

404
Chapter 17: Androgen therapy in non-gonadal disease

Sardar P, Jha A, Roy D, Majumdar U, testosterone gel (AndroGel) versus Sloane DE, Lee CW, Sheffer AL (2007)
Guha P, Roy S, Banerjee R, Banerjee intramuscular testosterone in Hereditary angioedema: safety of
AK, Bandyopadhyay D (2010) testosterone-deficient HIV-infected long-term stanozolol therapy.
Therapeutic effects of nandrolone men. HIV Clin Trials 8:412420 J Allergy Clin Immunol 120:654658
and testosterone in adult male HIV Seidman SN, Orr G, Raviv G, Levi R, Snyder PJ, Peachey H, Hannoush P,
patients with AIDS wasting Roose SP, Kravitz E, Amiaz R, Berlin JA, Loh L, Lenrow DA,
syndrome (AWS): a randomized, Weiser M (2009) Effects of Holmes JH, Dlewati A, Santanna J,
double-blind, placebo-controlled testosterone replacement in Rosen CJ, Strom BL (1999) Effect of
trial. HIV Clin Trials 11:220229 middle-aged men with dysthymia: a testosterone treatment on body
Sattler FR, Jaque SV, Schroeder ET, randomized, placebo-controlled composition and muscle strength in
Olson C, Dube MP, Martinez C, clinical trial J Clin Psychopharmacol men over 65 years of age. J Clin
Briggs W, Horton R, Azen S (1999) 29:216221 Endocrinol Metab 84:26472653
Effects of pharmacological doses of Shahidi NT (1973) Androgens and Spaulding WB (1960)
nandrolone decanoate and erythropoiesis. N Engl J Med Methyltestosterone therapy for
progressive resistance training in 289:7280 hereditary episodic edema
immunodeficient patients infected (hereditary angioneurotic edema).
with human immunodeficiency Sharma S, Arneja A, McLean L,
Duerksen D, Leslie W, Sciberras D, Ann Intern Med 53:739745
virus. J Clin Endocrinol Metab
84:12681276 Lertzman M (2008) Anabolic Spiers ASD, DeVita SF, Allar MJ,
steroids in COPD: a review and Richards S, Sedranak N (1981)
Saylor PJ, Smith MR (2010) Adverse preliminary results of a randomized Beneficial effects of an anabolic
effects of androgen deprivation trial. Chron Respir Dis 5:169176 steroid during cytotoxic
therapy: defining the problem and chemotherapy for metatstatic
promoting health among men with Sheffer AL, Fearon DT, Austen KF
(1977) Methyltestosterone therapy cancer. J Med 12:433446
prostate cancer. J Natl Compr Canc
Netw 8:211223 in hereditary angioedema. Ann Srinivas-Shankar U, Roberts SA,
Intern Med 86:306308 Connolly MJ, OConnell MD,
Schneider BK, Pickett CK, Zwillich Adams JE, Oldham JA, Wu FC
Sicotte NL, Giesser BS, Tandon V,
CW, Weil JV, McDermott MT, (2010) Effects of testosterone on
Klutch R, Steiner B, Drain AE,
Santen RJ, Varano LA, White DP muscle strength, physical function,
Shattuck DW, Hull L, Wang HJ,
(1986) Influence of testosterone on body composition, and quality of
Elashoff RM, Swerdloff RS, Voskuhl
breathing during sleep. J Appl life in intermediate-frail and frail
RR (2007) Testosterone treatment
Physiol 61:618623 elderly men: a randomized, double-
in multiple sclerosis: a pilot study.
Schofield RS, Hill JA, McGinn CJ, Arch Neurol 64:683688 blind, placebo-controlled study.
Aranda JM (2002) Hormone J Clin Endocrinol Metab 95:639650
Siddique H, Smith JC, Corrall RJ
therapy in men and risk of cardiac Stenner J, Holthaus K, Mackenzie SH,
(2004) Reversal of polycythaemia
allograft rejection. J Heart Lung Crawford ED (1998) The effect of
induced by intramuscular androgen
Transplant 21:493495 ejaculation on prostate-specific
replacement using transdermal
Schols AM, Soeters PB, Mostert R, testosterone therapy. Clin antigen in a prostate cancer-
Pluymers RJ, Wouters EF (1995) Endocrinol (Oxf) 60:143145 screening population. Urology
Physiologic effects of nutritional 51:455459
Silverberg D, Wexler D, Blum M,
support and anabolic steroids in Strawford A, Barbieri T, Van Loan M,
Schwartz D, Iaina A (2004) The use
patients with chronic obstructive of androgens in anaemia resistant to Parks E, Catlin D, Barton N, Neese
pulmonary disease. A placebo- R, Christiansen M, King J,
erythropoietin and i.v. iron in
controlled randomized trial. patients with heart and renal Hellerstein MK (1999) Resistance
Am J Respir Crit Care Med exercise and supraphysiologic
failure. Nephrol Dial Transplant
152:12681274 19:1021 androgen therapy in eugonadal men
Schols AM, Slangen J, Volovics L, Singh AB, Norris K, Modi N, with HIV-related weight loss: a
Wouters EF (1998) Weight loss is a Sinha-Hikim I, Shen R, Davidson T, randomized controlled trial. JAMA
reversible factor in the prognosis of Bhasin S (2001) Pharmacokinetics 281:12821290
chronic obstructive pulmonary of a transdermal testosterone system Sullivan DH, Roberson PK, Johnson
disease. Am J Respir Crit Care Med in men with end stage renal disease LE, Bishara O, Evans WJ, Smith ES,
157:17911797 receiving maintenance hemodialysis Price JA (2005) Effects of muscle
Scott JD, Wolfe PR, Anderson P, and healthy hypogonadal men. strength training and testosterone in
Cohan GR, Scarsella A (2007) J Clin Endocrinol Metab frail elderly males. Med Sci Sports
Prospective study of topical 86:24372445 Exerc 37:16641672

405
Chapter 17: Androgen therapy in non-gonadal disease

Svartberg J (2010) Androgens and Group) (1967) Carcinoma of the von Hartitzsch B, Kerr DNS (1976)
chronic obstructive pulmonary prostate: treatment comparisons. Response to parenteral iron with
disease. Curr Opin Endocrinol J Urol 98:516522 and without androgen therapy in
Diabetes Obes 17:257261 van Coevorden A, Stolear JC, Dhaene patients undergoing regular
M, van Herweghem JL, Mockel J haemodialysis. Nephron 17:430438
Svartberg J, Aasebo U, Hjalmarsen A,
Sundsfjord J, Jorde R (2004) (1986) Effect of chronic oral Walters KA, Simanainen U,
Testosterone treatment improves testosterone undecanoate Handelsman DJ (2010) Molecular
body composition and sexual administration on the pituitary- insights into androgen actions in
function in men with COPD, in a 6- testicular axes of hemodialyzed male and female reproductive
month randomized controlled trial. male patients. Clin Nephrol function from androgen receptor
Respir Med 98:906913 26:4854 knockout models. Hum Reprod
van den Berghe G, Wouters P, Weekers Update 16:543558
Talbot JA, Sheldrick R, Caswell H,
Duncan S (2008) Sexual function in F, Verwaest C, Bruyninckx F, Schetz Warren MF, Serby MJ, Roane DM
men with epilepsy: how important is M, Vlasselaers D, Ferdinande P, (2008) The effects of testosterone on
testosterone? Neurology 70:13461352 Lauwers P, Bouillon R (2001) cognition in elderly men: a review.
Intensive insulin therapy in the CNS Spectr 13:887897
Teruel JL, Marcen R, Navarro JF, surgical intensive care unit. N Engl
Villafruela JJ, Fernandez-Lucas M, Watson RN, Bradley MH, Callahan R,
J Med 345:13591367 Peters BJ, Kory RC (1959)
Liano F, Ortuno J (1995) Evolution
of serum erythropoietin after Van Vollenhoven RF, Engleman EG, A six-month evaluation of an
androgen administration to McGuire JL (1995) anabolic drug, norethandrolone, in
hemodialysis patients: a prospective Dehydroepiandrosterone in underweight persons. Am J Med
study. Nephron 70:282286 systemic lupus erythematosus. 26:238248
Arthritis Rheum 38:18261831 Wattel E, Cambier N, Caulier MT,
Teruel JL, Aguilera A, Marcen R,
Antolin JN, Otero GG, Ortuno J Vaughan C, Goldstein FC, Tenover JL Sautiere D, Bauters F, Fenaux P
(1996a) Androgen therapy for (2007) Exogenous testosterone (1994) Androgen therapy in
anaemia of chronic renal failure. alone or with finasteride does not myelodysplastic syndromes with
Scand J Urol Nephr 30:403408 improve measurements of cognition thrombocytopenia: a report
in healthy older men with low on 20 cases. Br J Haematol
Teruel JL, Marcen R, Navarro-Antolin serum testosterone. J Androl 87:205208
J, Aguilera A, Fernandez-Juarez G, 28:875882
Ortuno J (1996b) Androgen versus Webb WR, Doyle RS, Howard HS
erythropoietin for the treatment of Vecchio M, Navaneethan SD, Johnson (1960) Relative metabolic effects of
anemia in hemodialyzed patients: a DW, Lucisano G, Graziano G, calories, protein, and an anabolic
prospective study. J Am Soc Nephrol Saglimbene V, Ruospo M, Querques steroid (19-nortestosterone) in early
7:140144 M, Jannini EA, Strippoli GF (2010) postoperative period. Metabolism
Interventions for treating sexual 9:10471057
Teruel JL, Cano T, Marcen R, dysfunction in patients with chronic
Villafruela JJ, Rivera M, Fernandez- Weber JP, Walsh PC, Peters CA,
kidney disease. Cochrane Database Spivak JL (1991) Effect of reversible
Juarez G, Ortuno J (1997) Decrease Syst Rev 12:CD007747
in the haemoglobin level in androgen deprivation on
haemodialysis patients undergoing Velazquez I, Alter BP (2004) hemoglobin and serum
antiandrogen therapy. Nephro Dial Androgens and liver tumors: erythropoietin in men. Am
Transplant 12:12621263 Fanconis anemia and non- J Hematol 36:190194
Fanconis conditions. Am J Hematol Wells R (1960) Prednisolone and
Thomas S, Wolf SE, Murphy KD, 77:257267 testosterone propionate in cirrhosis
Chinkes DL, Herndon DN (2004)
Victor G, Shanmugasundaram K, of the liver: a controlled trial. Lancet
The long-term effect of oxandrolone
Krishnamurthi CA, Rex PM, 2:14161419
on hepatic acute phase proteins in
Nagarajan D (1967) Haemoglobin Whitnall MH, Elliott TB, Harding RA,
severely burned children. J Trauma
response to anabolic steroid in iron-
56:3744 Inal CE, Landauer MR, Wilhelmsen
deficiency anaemia. J Assoc CL, McKinney L, Miner VL, Jackson
Tweedle D, Walton C, Johnston IDA Physicians India 15:177183 WE 3rd, Loria RM, Ledney GD,
(1973) The effect of an anabolic
Villareal DT, Morley JE (1994) Trophic Seed TM (2000) Androstenediol
steroid on postoperative nitrogen
factors in aging. Should older stimulates myelopoiesis and
balance. Br J Clin Pract 27:130132
people receive hormonal enhances resistance to infection in
VACURG (Veterans Administration replacement therapy? Drugs Aging gamma-irradiated mice. Int
Cooperative Urological Research 4:492509 J Immunopharmacol 22:114

406
Chapter 17: Androgen therapy in non-gonadal disease

Wilkey JL, Barson LJ, Kest L, Bragagni Wu FC, von Eckardstein A (2003) Harmon D, Hewlett J, Fay J,
A (1960) The effect of testosterone Androgens and coronary Mangan K (1988) A multicenter
on the azotemic patient: an artery disease. Endocr Rev trial of antithymocyte globulin in
intermediary report. J Urol 24:183217 aplastic anemia and related diseases.
83:2529 Wu FCW, Farley TMM, Peregoudov A, Blood 72:18611869
Williams JS, Stein JH, Ferris TF (1974) Waites GMH; WHO Task Force on Young RL, Fuchs RJ, Woltjen MJ
Nandrolone decanoate therapy for Methods for the Regulation of Male (1976) Chorionic gonadotropin in
patients receiving hemodialysis. Fertility (1996) Effects of weight control. A double-blind
A controlled study. Arch Intern Med testosterone enanthate in normal crossover study. JAMA
134:289292 men: experience from a multicenter 236:24952497
Winearls CG (1995) Historical review contraceptive efficacy study. Fertil
Zak PJ, Kurzban R, Ahmadi S,
of the use of recombinant human Steril 65:626636
Swerdloff RS, Park J, Efremidze L,
erythropoietin in chronic renal Wulfsohn NL, Politzer WM, Henrico Redwine K, Morgan K, Matzner W
failure. Nephrol Dial Transplant 10 JS (1964) Testosterone therapy in (2009) Testosterone administration
(Suppl 2):39 bronchial asthma. S Afr Med J decreases generosity in the
Wolf SE, Edelman LS, Kemalyan N, 38:170172 ultimatum game. PLoS One 4:e8330
Donison L, Cross J, Underwood M, Young GA, Yule AG, Hill GL (1983) Zarrouf FA, Artz S, Griffith J, Sirbu C,
Spence RJ, Noppenberger D, Effects of an anabolic steroid on Kommor M (2009) Testosterone
Palmieri TL, Greenhalgh DG, plasma amino acids, proteins, and and depression: systematic review
Lawless M, Voigt D, Edwards P, body composition in patients and meta-analysis. J Psychiatr Pract
Warner P, Kagan R, Hatfield S, Jeng receiving intravenous 15:289305
J, Crean D, Hunt J, Purdue G, hyperalimentation. J Parent Ent
Burris A, Cairns B, Kessler M, Klein Nutr 7:221225 Zawilska K, Tokarz A, Misiak A, Psuja
RL, Baker R, Yowler C, Tutulo W, P, Wislawski S, Szymczak P,
Young LA, Neiss MB, Samuels MH, Meissner J, Karon J, Lewandowski
Foster K, Caruso D, Hildebrand B,
Roselli CE, Janowsky JS (2010) K, Lopaciuk S, Ziemski JM, Sowler J
Benjamin W, Villarreal C, Sanford
Cognition is not modified by large (1990) Nebulised heparin and
AP, Saffle J (2006) Effects of
but temporary changes in sex anabolic steroid in the prevention of
oxandrolone on outcome measures
hormones in men. J Clin Endocrinol postoperative deep venous
in the severely burned: a multicenter
Metab 95:280288 thrombosis following elective
prospective randomized double-
blind trial. J Burn Care Res Young N, Griffith P, Brittain E, abdominal surgery. Folia
27:131139; discussion:140141 Elfenbein G, Gardner F, Huang A, Haematologica 117:699707

407
Chapter
Review of guidelines on diagnosis and

18 treatment of testosterone deficiency


Ronald S. Swerdloff and Christina C. L. Wang

18.1 Introduction 408 18.5.3 Muscle strength and function 415


18.2 What is a guideline? 408 18.5.4 Bone mineral loss and
18.2.1 Definition of guideline 408 osteoporosis 415
18.2.2 Selection of guideline committee 18.5.5 Cognition 416
members 413 18.5.6 Mood and well-being 416
18.2.3 Evidence-based versus expert-opinion 18.5.7 Diabetes and metabolic syndrome 416
criteria for guidelines 413 18.6 Potential risks of testosterone treatment 416
18.3 Definition of hypogonadism 413 18.6.1 Prostate cancer 416
18.3.1 Diagnosis of hypogonadism 413 18.6.2 Severe lower urinary tract obstructive
18.3.2 Age-group selective? 413 symptoms (LUTS) and benign prostate
18.3.3 Hormone threshold 414 hyperplasia 416
18.3.4 What level of testosterone is clinically 18.6.3 Cardiovascular disease 416
relevant for the diagnosis? 414 18.6.4 Erythrocytosis 417
18.3.5 Symptoms required for the diagnosis of 18.6.5 Untreated sleep apnea 417
hypogonadism 414 18.7 Treatment options 417
18.4 Screening for androgen deficiency 18.8 Monitoring patients on testosterone
syndrome 414 treatment 417
18.5 Clinical manifestations of 18.9 Conclusions 417
hypogonadism 414 18.10 Key messages 418
18.5.1 Libido 414 18.11 References 419
18.5.2 Erectile function 415

18.1 Introduction particular processes according to a set routine or


This is an effort to review and compare the guidelines sound practice. By definition, following a guideline
on diagnosis, treatment options, outcomes and moni- is never mandatory. Guidelines are thus not binding
toring of men with low serum testosterone. The prin- and should not be enforced. Medical guidelines are
cipal published guidelines are listed in Table 18.1. often created by medical professional societies to
help physicians and other health professionals pro-
vide community-level standard of practice, and are
18.2 What is a guideline? sometimes used to reward practitioners for desired
18.2.1 Definition of guideline behavior.
A guideline is a statement by which to determine a
course of action. A guideline aims to streamline

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

408
Table 18.1 Comparison of recent guidelines on management of male hypogonadism

Organization Applicable Diagnosis: Screening Treatment Serum Exclusions Monitoring


Title age symptoms, options testosterone requirements for
Date of newest hormone tests and listed goals testosterone
guideline threshold treatment
American All ages; Symptoms: desirable Not stated Many Mid-normal Symptoms: every 3
Association of discusses Hormones: 4 months in yr 1
Clinical LOH TT: multiple <200 serum T:
Endocrinologists 318 ng/dl (<6.9 to recommended but
(Petak et al. 2002) 11 nmol/l) frequency not stated
Medical guidelines Repeat TT required: DRE and prostate
for clinical not stated symptoms: every 612
practice for the SHBG: can be used to months; PSA annually
evaluation and calculate FT HCT: every 6 months
treatment of FT, BT, cFT: optional; for 18 months, and
hypogonadism in no thresholds annually; T treatment
adult male provided adjusted if HCT>50%
patients Lipids: initial; 612
2002 months and annually
Not evidence
based
Swedish All ages Symptoms: required No Yes Not stated DRE abnormal; PSA > T, PSA, hemoglobin,
specialists Hormones: 3 ng/l; breast CA; HCT HCT: every 3 months
(Abrahamsson TT: <8 nmol/l (230ng/ > 53% (relative); OSA for 1 yr then every
et al. 2010) dl) probable (relative); heart failure 12 months
Testosterone deficiency; >12 nmol/l (untreated) DRE: every 12 months
deficiency (346 nmol/l) probably BMD: every 24 months
management normal; 812 nmol/l Lipids: not stated
program and gray area
treatment Repeat TT required:
recommendations not stated
Unpublished SHBG: yes
Not evidence FT, BT, cFT: no
based
Canadian Older age Symptoms: required Only high risk Yes Not stated Breast or prostate CA; Serum T: every 36
Physicians group > Hormones: men; abnormal PSA and months in year 1 and
(Morales et al. age 40 TT: no threshold challenges DRE refer to urologist; yearly thereafter
2010) specificity of IPSS > 21 relative
Table 18.1 (cont.)

Organization Applicable Diagnosis: Screening Treatment Serum Exclusions Monitoring


Title age symptoms, options testosterone requirements for
Date of newest hormone tests and listed goals testosterone
guideline threshold treatment
Practical guide to Repeat TT required: ADAM contraindication to PSA and DRE: every
diagnosis, yes questionnaire T treatment; 36 months in year 1
management and SHBG: only as part of erythrocytosis; and at yearly intervals
treatment of cFT and cBT; no untreated OSA; severe Hemoglobin and HCT:
testosterone threshold levels CHF; men wishing every 36 months in
deficiency for given biological fatherhood year 1 and yearly
Canadian FT, BT, cFT: BT gold thereafter
physicians standard; cBT and Lipids: not stated
2010 cFT acceptable
alternative; no
threshold levels
given
Endocrine Society Young and Symptoms: required No, except in Yes Mid-normal Prostate CA; breast CA; Serum T: 36 months
(Bhasin et al. 2010) middle age Hormones: certain high- (young and PSA > 4.0 ng/ml and/ after starting T Rx
Testosterone men; older TT: for young and risk middle-aged or abnormal DRE (>3.0 PSA/DRE: no for age
therapy in men men middle-aged men, populations men); older PSA in high-risk e.g. < 40; >40 baseline
with androgen equivocal below reference range (e.g. HIV and men unclear African and first- DRE and PSA, 36
deficiency for healthy young glucocorticoid- degree relatives with months after T Rx,
syndromes: an men established in receiving men) prostate CA: refer to then guidelines for
Endocrine Society individual laboratory. urologist; suggest age and race; increase
clinical practice For older men, panel prostate CA risk in PSA > 1.4 ng/ml
guideline disagreed on calculator be used. within any 12-month
2010 threshold HCT >50%; IPSS >19; period or PSA velocity
Evidence based TT: <280 ng/dl hyper-viscosity, > 0.4 ng/ml per yr
(9.7 nmol/l) with untreated OSA; using PSA level after 6
symptoms or <200 class IIIIV CHF months of T Rx as
ng/dl (6.9 nmol/l) reference and PSA
Repeat TT required: data available for
yes >2years; IPSS > 19
SHBG: no, unless cFT HCT: baseline 36
or BT are used. months and annual
Table 18.1 (cont.)

Organization Applicable Diagnosis: Screening Treatment Serum Exclusions Monitoring


Title age symptoms, options testosterone requirements for
Date of newest hormone tests and listed goals testosterone
guideline threshold treatment
FT, BT, cFT: no, except >54%: stop therapy
when TT is in gray and adjust dose
zone or altered SHBG Lipids: not stated
suspected
ISA, ISSAM, EAU, > age 60 Symptoms: not stated, No Yes Mid- to lower Prostate and breast Serum T: not stated
EAA, ASA improvement in signs young adult CA; erythrocytosis; DRE and PSA: baseline,
(Wang et al. 2009) and symptoms of male serum (HCT > 52%) at 36 months, 12
Investigation, T deficiency should be T levels untreated; OSA months and annually
treatment and sought by 36 months untreated; severe CHF; after Rx; suspicious
monitoring of of treatment IPSS > 21 relative DRE/increased PSA or
late-onset Hormones: contraindication to calculated risk leads
hypogonadism in TT: <8 nmol/l (230 ng/ T treatment to ultrasound-guided
males dl); >12 nmol/l biopsy; men
2010 (350 ng/dl) not successfully treated
Evidence based hypogonadal; 812 for prostate CA with
nmol/l (230350 ng/ symptomatic
dl) requires calculated hypogonadism are
FT or FT by equilibrium potential candidates
dialysis for T Rx after a prudent
Repeat TT required: interval if there is no
not stated, monitor clinical or laboratory
improvement in signs evidence of residual
and symptoms of CA
T deficiency by 36 HCT/hemoglobin:
months of treatment before Rx, 34 months
SHBG: required for and 12 months of Rx
calculated FT then annually; dose
FT, BT, cFT: FT <225 adjustment or periodic
pmol/l (65 pg/ml) phlebotomy to keep
provides supportive hematocrit below 55%
evidence for Lipids: not stated
T treatment; threshold
values for BT are not
generally available;
salivary T not
recommended for
general use
Table 18.1 (cont.)

Organization Applicable Diagnosis: Screening Treatment Serum Exclusions Monitoring


Title age symptoms, options testosterone requirements for
Date of newest hormone tests and listed goals testosterone
guideline threshold treatment
Practice > age 50 Symptoms: required No Yes Not stated Clinical prostate CA; Serum T: levels over
Committee for the Hormones: breast CA; elevated pretreatment but not
American Society TT: < 200 ng/dl HCT > 55%; and >reference range for
for Reproductive (<6.9 nmol/l) = sensitivity to young adult males
Medicine hypogonadism; >400 treatment; relative PSA: PSA >1.0 ng/ml
(American Society (13.8 nmol/l) not contraindications are: within 36 months of
for Reproductive deficient; 200400 severe OSA; HCT> Rx; PSA obtained 3
Medicine 2006) treatment could 52%; severe LUTS; and and 6 months after
Treatment of be beneficial CHF treatment and
androgen Repeat TT required: annually thereafter
deficiency in the not stated HCT, hemoglobin: at
aging male SHBG: yes for cFT or 3 and 6 months after
2006 cBT Rx and annually
Not evidence FT, BT, cFT: FT or BT or Lipids: not stated
based cFT may be useful for
might benefit group;
values for BT that are
<normal range for
adult men supports
the diagnosis of
T deficiency
Andrology All ages Symptoms: required? Not stated Yes Not stated prostate and breast Not stated
Australia Hormones: CA; relative exclusion
(www. TT: <8 nmol/l (230 ng/ men with OSA and
andrologyaustralia. dl) CHF
org) Repeat TT required:
Testosterone not stated
deficiency fact SHBG: no, unless as
sheet alternative if cFT or BT
2005 are used
Patient FT, BT, cFT: not stated
information sheet
Abbreviations: T, testosterone; TT, total testosterone; FT, free testosterone; BT, bioavailable testosterone; cFT, calculated FT; cBT, calculated BT; DRE, digital rectal examination;
PSA, prostate-specific antigen; LUTS, lower urinary tract symptoms; HCT, hematocrit; OSA, obstructive sleep apnea; CHF, chronic heart failure; IPSS, International Prostate Symptom Score;
Rx, treatment; CA, cancer; ISA, International Society of Andrology; ISSAM, International Society for the Study of the Aging Male; EAU, European Association of Urology; EAA, European
Acadamy of Andrology; ASA, American Society of Andrology.
Chapter 18: Guidelines for diagnosis and treatment

18.2.2 Selection of guideline committee American Association of Clinical Endocrinologists


(Petak et al. 2002), Swedish Specialists
members (Abrahamsson et al. 2010), the Endocrine Society of
Guideline committee members are usually selected by Australia (Conway et al. 2000) and American
a professional organization because they are experi- Society of Reproductive Medicine (American
enced and respected in the particular area of interest. Society for Reproductive Medicine 2006) fall
In some cases guidelines can be written by special- into this category.
interest groups with predetermined agendas. Organization-based or other: many guidelines are
identified as an official position of a medical or
18.2.3 Evidence-based versus scientific society, while others appear to be an
independent select group of experts. Some
expert-opinion criteria for guidelines guidelines seem to be sponsored by a
Guidelines can be based on expert opinion or on pub- pharmaceutical company. In this last situation it
lished evidence at defined levels of quality (evidence may not be clear if the recommendations were
based). The highest quality for interventional studies is fully independent.
appropriately powered, double-blinded and placebo-
controlled clinical trials. Unfortunately, many guide-
lines lack adequate high levels of evidence, and thus 18.3 Definition of hypogonadism
such guidelines are to a great extent expert-opinion Hypogonadism in the literal sense refers to a decreased
formulated. This is the case for male hypogonadism. function of the hormone secretory compartments of
In this review, the authors have surveyed a number the testes (e.g. testosterone secretion by Leydig cells)
of guidelines on androgen deficiency, and present the and decreased sperm production by the germinal
major consensus views as well as areas where there compartment. However, most often clinicians think of
are significant differences in opinions. The guidelines hypogonadism as androgen deficiency and its clinical
are listed in Table 18.1. It will be noted that some manifestations. This review of hypogonadism guide-
guidelines focus on all age ranges and are independent lines will focus on testosterone deficiency.
of the etiologies of hypogonadism, while others focus
on specific age groups (i.e. old age), etiologies (i.e.
HIV, obesity), targeted symptoms (i.e. libido, osteo-
18.3.1 Diagnosis of hypogonadism
porosis) and outcomes. Some focus on different inter- The diagnosis of testosterone deficiency may be based
ventional approaches (testosterone treatment). only on the blood hormone level (serum testosterone)
or require both a low serum testosterone and symp-
Evidence-based: the International Societies (Wang
toms compatible with, or specific for, a low serum
et al. 2009) and The Endocrine Society (Bhasin et al.
testosterone. Unfortunately, many symptoms lack
2010) guidelines rated the quality of the evidence
sufficient specificity to be diagnostic for a low serum
and thus can be considered evidence based,
testosterone. Furthermore, blood levels of testoster-
although both acknowledged that much of the
one can be assayed as total testosterone, free testoster-
evidence is weak and the conclusions are thus to
one or biologically active testosterone. Each type of
some extent expert opinion. The authors of this
measurement uses different methodologies which
chapter have participated on several of the
may vary in accuracy and specificity.
guidelines committees referenced in this review,
and can attest to the fact that there may be
differences in the views of guideline committee 18.3.2 Age-group selective?
members on specific areas; recommendations may Testosterone deficiency in males differs in diagnostic
represent a negotiated consensus view. approaches and clinical presentations in infants, chil-
Expert opinion: a number of the reviewed guidelines dren, pubertal, young and middle-aged adults and in
provided opinions that were not rated by the quality old age. This review focuses on adults. Most guide-
of evidence. All guidelines provided literature lines are broad in reference to age group, but some
support for what could be considered expert have specifically dealt with androgen deficiency in the
opinions. The guidelines on hypogonadism from elderly, referred to in some guidelines as late onset
the Canadian Physicians (Morales et al. 2010), hypogonadism (LOH) (Wang et al. 2009).

413
Chapter 18: Guidelines for diagnosis and treatment

18.3.3 Hormone threshold while others insist on both a chemical deficiency


and clinical signs or symptoms to merit the diagnosis
This is one of the most difficult aspects of defining hypo-
of hypogonadism (Table 18.1).
gonadism. The standard approach is to take a reference
range for a normal population (mean  2 SD) and define
hypogonadism as below this population-based standard. 18.4 Screening for androgen
This approach has several problems: (1) What popula- deficiency syndrome
tion? (Age, race, exclusions for co-morbidities.) (2) What
reference range? Unfortunately we lack a universally Very few guidelines support the use of either population-
agreed-upon reference range (published reference ranges based hormonal screening or questionnaires to find
vary considerably depending on the laboratory method- more cases of hypogonadism (Table 18.1; Wang
ology and population studied). (3) What testosterone et al. 2009; Bhasin et al. 2010). Guideline authors seem
assays used? For example, total testosterone, biologically to be inhibited by the lack of clear hormone thresholds
active testosterone, and free testosterone. Free testos- (see above) and limited evidence-based data to prove
terone can be measured by analog displacement assays, benefit for treatment of the full spectrum of symptoms,
calculated based on formulae of mass action or empirical to support the use of population-based testosterone
formulae (Sartorius et al. 2009; Ly et al. 2010), assessed measurement screening. The case-finding question-
knowing SHBG and total testosterone levels, or deter- naires such as Androgen Deficiency in Aging Male
mined by equilibrium dialysis (Vermeulen et al. 1999). (ADAM) (Morley et al. 2000), Aging Male Symptoms
(See Chapter 4.) Rating Scale (AMS) (Heinemann et al. 2004) and the
Massachusetts Male Aging Study Questionnaire (Smith
18.3.4 What level of testosterone is et al. 2000) proposed for identification of hypogonad-
ism are limited in practical use by the lack of specificity
clinically relevant for the diagnosis? (despite great sensitivity). There is a new questionnaire
There have been efforts to relate symptoms and signs of for hypogonadal men that has not yet been validated in
hypogonadism to serum testosterone levels using defined different populations (Rosen et al. 2011). Thus, essen-
methodology. This is best exemplified by the data from tially none of the guidelines have advocated population-
the European Male Aging Study (Wu et al. 2010). These based screening, although some have advocated use of
data suggest that different symptom components of hypo- hormone screening in high-risk groups. For example,
gonadism seem to present at different serum testosterone the Endocrine Society supports measuring serum
concentrations in a population. Thus, decreased libido is testosterone in patients receiving opioids and glucocor-
evident at a higher serum testosterone level than other ticoids and men with known HIV disease, and low
symptoms. Different guidelines create a threshold from trauma fractures, regardless of symptoms of low testos-
anywhere below a defined reference range to levels below terone; while they recommend measurement of serum
200 ng/dl. In some guidelines, expert opinions were testosterone in men with type 2 diabetes mellitus,
acknowledged to differ considerably (Wang et al. 2009). chronic renal disease and chronic obstructive pulmon-
The problem is further complicated by the concept that ary disease only when they have symptoms suggestive of
there are individual patient differences in sensitivity to low serum testosterone. A similar recommendation was
serum testosterone. This concept is inherent in the range stated for type 2 diabetes by the International Societies
of values that make up the bell-shaped curve of a reference guidelines; the American Association of Clinical Endo-
population. Thus a normal man, A, may have a mean crinologists (AACE) guidelines were silent on this topic.
serum testosterone of 600 ng/dl, while equally normal
man, B, may appear phenotypically indistinguishable in
terms of signs and symptoms of androgen effect from a 18.5 Clinical manifestations of
man with a mean serum testosterone of 400 ng/dl. hypogonadism
18.3.5 Symptoms required for the 18.5.1 Libido
Impaired libido is the classic symptom of low testos-
diagnosis of hypogonadism terone and is a focal point of the symptom complex
Some guidelines refer only to objective evidence of a in the discussion of most guidelines. The EMAS
low serum testosterone to define hypogonadism, defined the serum testosterone level that is associated

414
Chapter 18: Guidelines for diagnosis and treatment

with low testosterone syndrome in middle-aged and in men with low testosterone (American Society
elderly men (Wu et al. 2010). Impaired libido is also for Reproductive Medicine 2006; Wang et al. 2009;
noted in LOH in several guidelines (Wang et al. 2009; Bhasin et al. 2010; Morales et al. 2010). Most but not
Bhasin et al. 2010; Morales et al. 2010). The AACE all guidelines discuss improved muscle mass with
guidelines of 2002 stated that most studies support the testosterone treatment and emphasize variability of
benefits of testosterone therapy for impaired sexual improvement in function. Meta-analyses of random-
behavior in men with low testosterone (Petak et al. ized trials in middle-aged men and placebo-controlled
2002). The ISA guidelines recommend that men with studies in older men have demonstrated that TRT is
ED and/or decreased libido and documented low associated with greater improvement in grip strength
testosterone are candidates for testosterone therapy and lean body mass and reduction in body fat mass
(Wang et al. 2009). The Canadian Physicians guide- than placebo (Isidori et al. 2005; Page et al. 2005;
lines are positive about the benefits of testosterone Srinivas-Shankar et al. 2010). In contrast, changes in
therapy for men with decreased sexual desire (Morales performance-based measures of physical function in
et al. 2010). The Endocrine Society guidelines recom- testosterone trials that recruited healthy older men
mend that clinicians offer testosterone therapy to selectively have been inconsistent across trials (Bhasin
men with low serum testosterone levels and low libido et al. 2010). It is hoped that the ongoing, larger sized,
and men with ED after evaluating for other causes of well-controlled testosterone interventional trials on
ED and consideration of other therapies for ED. They improvement in physical function in older men with
go on to state that the evidence that testosterone is physical disabilities will provide more clarity to the
beneficial for improving decreased libido is overall important question of reduction in frailty after testos-
supportive but not always consistent across controlled terone replacement.
studies reported in the literature (Bhasin et al. 2010).

18.5.2 Erectile function 18.5.4 Bone mineral loss and osteoporosis


Most trials have recognized that low testosterone
Erectile dysfunction is also a classic symptom of
is a risk factor for decreased BMD and fractures
men with low testosterone; unless the testosterone
(especially in hypogonadal older men). Most experts
deficiency is very severe, it is often secondary to a
agree that the prevalence of osteoporosis is greater in
co-morbid condition that responds better to phos-
men with low, compared to normal, serum testoster-
phodiesterase 5 (PDE5) inhibitors than testosterone
one levels (Amin et al. 2000; Meier et al. 2008).
replacement. The ISA guideline on LOH discusses the
Testosterone replacement therapy was found to
possibility that testosterone treatment may be useful
increase bone density in hypogonadal men (Behre
as a complementary treatment when older hypogona-
et al. 1997; Svartberg et al. 2008). While a few studies
dal patients fail to respond to PDE5 inhibitors (Wang
have not shown a clear benefit in bone density, many
et al. 2009). The Canadian Physicians guideline states
studies with exogenous testosterone have noted
that men with profound hypogonadism show signifi-
increases in BMD in hypogonadal aging males (Merza
cant improvement in ED when on TRT (Morales et al.
et al. 2006). Some studies have reported improve-
2010). For ED, the use of PDE5 inhibitors remains the
ments in lumbar bone density (Snyder et al. 1999;
principal treatment. In men with low testosterone and
Amory et al. 2004; Wang et al. 2004; Basurto et al.
ED, the majority of experts seem to prefer initial
2008; Svartberg et al. 2008). This has also been noted
treatment with PDE5 inhibitors and adding testoster-
in two meta-analyses (Isidori et al. 2005; Tracz et al.
one for decreased libido or other signs and symptoms
2006). Some studies (Amory et al. 2004; Wang et al.
of low testosterone. A properly powered study is
2004; Nair et al. 2006) suggested improvements in hip
needed to compare the benefits of PDE5 inhibitors
bone mineral density; while meta-analysis studies
and testosterone, alone and together, in men with ED
(Isidori et al. 2005; Tracz et al. 2006) found femoral
and low testosterone.
neck improvements to be inconclusive. The Endo-
crine Society guidelines state that the available data
18.5.3 Muscle strength and function rule out a moderate to large testosterone effect on
A number of guidelines list decreased muscle mass BMD; yet some studies have shown impressive
and strength as one of the symptom complexes seen increases in BMD in older men (Amory et al. 2004).

415
Chapter 18: Guidelines for diagnosis and treatment

The Canadian Physicians guidelines say that bone levels in cases of metabolic syndrome or type 2 dia-
mineral density is increased by testosterone therapy in betes, insulin resistance is reduced and this appears to
men with low testosterone levels (Morales et al. improve both blood glucose control and dyslipidemia.
2010). American Association of Clinical Endocrinolo-
gists guidelines state that BMD should be assessed
before and at one to two years; treatment options to
18.6 Potential risks of testosterone
maintain bone mass may include testosterone, imply- treatment
ing that it is prescribed for other manifestations of
hypogonadism (Petak et al. 2002). The ISA guidelines
18.6.1 Prostate cancer
argue that bone density increases with testosterone All guidelines and regulatory-agency-directed medi-
treatment in hypogonadal men of all ages, but recog- cine instructions list non-cured prostate cancer as a
nizes that the lack of data on reduction of fractures contraindication for testosterone treatment. Paradox-
makes conclusions on the benefit of testosterone ically, most guidelines indicate that there was insuffi-
to treatment of hypogonadal men with low BMD cient data to determine if long-term testosterone is a
difficult (Wang et al. 2009). It is clear from the review risk factor for developing prostate cancer in the
of guidelines that prevention of fractures has not been future. The Endocrine Society guideline recommends
adequately assessed, and assessment of the long-term against testosterone therapy without further urologi-
benefits of testosterone on bone strength and density cal evaluation in men with a palpable prostate nodule
requires larger-sized, well-controlled studies. or induration. They made the same cautious state-
ment in men with PSA > 4 ng/ml or > 3 ng/ml in
men with high family risk or African racial derivation
18.5.5 Cognition (Bhasin et al. 2010).
There are very limited data on the effects of testosterone
therapy in hypogonadal men; the Endocrine Society
guidelines state that there were three placebo-controlled 18.6.2 Severe lower urinary tract
studies, but the results were imprecise on several obstructive symptoms (LUTS) and benign
dimensions of cognition and were insignificant after
pooling (Bhasin et al. 2010).
prostate hyperplasia
This widely used symptom list is often given in guide-
lines as a relative or absolute contraindication for
18.5.6 Mood and well-being testosterone treatment (Wang et al. 2009; Bhasin
Mild depressive states are reported in many hypogo- et al. 2010); yet there is little evidence that symptoms
nadal men. The Canadian Physician guideline states are worsened with treatment. Despite this, some
that testosterone therapy may benefit emotional well- guidelines give contraindication thresholds for the
being (Morales et al. 2010). The Endocrine Society International Prostate Symptom Score questionnaire
guidelines reviewed the interventional data and (IPSS) >19 or 21, as examples.
found that results of controlled studies on depression
and QoL were imprecise and inconsistent (Bhasin
et al. 2010).
18.6.3 Cardiovascular disease
Cardiovascular risk is discussed in many guidelines.
The principal position is that low testosterone is asso-
18.5.7 Diabetes and metabolic syndrome ciated with greater cardiovascular morbidity and
There are many studies suggesting that low serum mortality, but there is inadequate interventional data
testosterone may be a predictor of future metabolic to say whether testosterone treatment of men with
syndrome and diabetes mellitus (Brand et al. 2010; low testosterone will benefit cardiovascular status,
Wang et al. 2011). Evidence that interventional worsen or not affect cardiovascular outcomes. The
replacement testosterone treatment will prevent or cor- Swedish Specialists guidelines (Abrahamsson et al.
rect either metabolic syndrome or type 2 diabetes 2010) are more adamant that there is no increased
mellitus is limited. The Swedish Specialists (Abrahams- risk from testosterone treatment on coronary disease.
son et al. 2010) state, however, that hypothesis- They do, however, say that fluid retention induced by
generating studies show that, by normalizing testosterone testosterone may worsen CHF. This is also stated in

416
Chapter 18: Guidelines for diagnosis and treatment

other guidelines. A recent report of increased cardio- and adverse effects. It is unclear what testosterone
vascular risk of testosterone treatment of hypogonadal level should be a goal, for lack of benefit/risk data
debilitated men (Basaria et al. 2010; see Chapter 10) based on serum testosterone level attained. In general,
represents a potential cautionary sign, but the finding normal range levels are recommended. Safety blood
requires confirmation as it differs from other investi- testing includes PSA, hematocrit or hemoglobin;
gators who report cardiovascular benefits from testos- physical examination includes digital rectal exam
terone treatment (see Chapter 10). for prostate induration and nodules; history includes
lower urinary tract symptoms (LUTS) and obstructive
sleep apnea. The recommendation for frequency of
18.6.4 Erythrocytosis testing is not consistent among guidelines but, in
It was noted in most guidelines that testosterone will general, recommended at baseline, three to six
increase red blood cell mass, hemoglobin and hema- months and annually thereafter. Some guidelines pro-
tocrit. The Endocrine Society guidelines recommend vide detailed PSA and hematocrit levels (Wang et al.
against treating men with low testosterone if their 2009; Bhasin et al. 2010) for stopping treatment,
hematocrit is greater than 50% (Bhasin et al. 2010). evaluation with specialist or intervention (e.g. veni-
The ISA guidelines place the threshold for entry into section for erythrocytosis).
treatment at hematocrit of >52% (Wang et al. 2009),
and the Swedish Specialists guidelines (Abrahamsson
et al. 2010) recommend hematocrit >53% as a relative 18.9 Conclusions
contraindication The Canadian Physicians (Morales This chapter reviews a number of guidelines prepared
et al. 2010) and AACE (Petak et al. 2002) guidelines by experts in a number of countries in North America
do not define erythrocytosis, but list it as a contra- and Europe for the clinical diagnosis and treatment of
indication to treatment with testosterone. young adult, middle-aged and elderly hypogonadal
men. These guideline committees reviewed the litera-
18.6.5 Untreated sleep apnea ture on the topic to provide evidence-based and/or
This is listed as a contraindication or relative contra- expert opinion, with two guidelines attempting to
indication in most guidelines. The evidence for provide an assessment of quality of the evidence:
physiological doses of testosterone aggravating or the Endocrine Society (Bhasin et al. 2010) and Inter-
causing obstructive sleep apnea is very weak but is national Societies (Wang et al. 2009). These two
usually listed in regulatory inserts as a risk. guidelines concluded that the evidence to support
many of the recommendations was either weak or
reverted to expert opinion. McLachlan, in an editorial
18.7 Treatment options opinion on the 2010 Endocrine Society guidelines,
Most guidelines reviewed available testosterone formu- stated The evidence basis for informing clinical prac-
lations, with some providing details on pharmacoki- tice has developed only marginally; among the 45
netics and drug-specific adverse effects. The ISA new references cited from 2006, there are 7 small to
guidelines express concern about use of long-acting moderate sized randomized controlled trials and sev-
testosterone injectables for older men with LOH eral controlled studies of responses in young vs. older
because of the protracted half-life that would delay men; the remainder are commentaries, meta-analyses,
withdrawal from drug in case of adverse effect (Wang or epidemiological studies. With their intimate
et al. 2009). The formulation list is provided in many understanding of the knowledge gaps and as required
guideline documents, but, since it is the most recent, for writing summary guidelines, the authors [of this
we refer readers to Table 6 in The Endocrine Society guideline] have filled in the blanks with considered
2010 guidelines (Bhasin et al. 2010) and to Chapter 15. commentary. His editorial also commented on
the common use of vague terms that reflect the
lack of hard data and the shifting sands of opinion
18.8 Monitoring patients on (McLachlan 2010).
testosterone treatment A second editorial on the Endocrine Society 2010
Most guidelines recommend monitoring patients for guidelines update was provided by Bradley Anawalt.
improvements in symptoms, blood testosterone levels He reiterated that As with the original 2006

417
Chapter 18: Guidelines for diagnosis and treatment

guidelines, the evidence for the 2010 guidelines is frequency of monitoring are also mainly common
weak. On a rating scale of very low, low, moderate sense opinions based on clinical experience rather
and high quality, all the recommendations were than evidence based. There is a fairly strong opinion
based on evidence judged to be very low or low in most guidelines against population screening to
and the majority of the recommendations were detect the syndromic disorder of testosterone defi-
based on very low quality evidence (Anawalt ciency, as the rationale for treatment is not strongly
2010). He pointed out many controversial areas in science based.
the Endocrine Society guidelines (most addressed The careful review of the available guidelines also
by the guidelines authors themselves), including demonstrates the bias of the various committees
concern about the limited value of digital rectal members. There are the enthusiasts and naysayers
exams as a monitoring safety tool to detect prostate in the identification of potentially treatable patients.
cancer, the lack of consensus of the value of the It is our opinion that the divergences of opinion are
prostate calculator as a tool to integrate the data resolvable as we gather more hard data from carefully
on prostate risk, and the conservative recommen- powered, well-controlled, large-scale, interventional
dation of using a testosterone increase in PSA of studies in men of all ages with low-testosterone
>1.5 pg/mlyear as signal for a (urological) assess- disorder. Our bias is a cautious positive view that
ment (Wolf et al. 2010). There are some inconsist- we will better define who should and who should
encies in his argument as it follows references not be treated and, for the former category, determine
(Andriole et al. 2009; Schroder et al. 2009) as to if the benefits of replacement therapy outweigh
the limited value in reducing mortality rates by the risks.
screening for prostate cancer in the general popula-
tion. Anawalt argues for large-scale efficacy and
safety studies to answer important questions about 18.10 Key messages
benefit/risk of TRT for symptomatic men with low  Guidelines are guides designed to improve
testosterone. patient care and not enforceable rules.
The authors of this chapter are aware of an  Guidelines can be evidence based, represent
ongoing study (T trial) in 800 hypogonadal men, expert opinion, or a combination of evidence and
age  65 years, using a carefully crafted, multiple opinion.
end-point, placebo-controlled, double-blinded study  Evidence-based guidelines are valued more than
design. The study will provide insights into the those based on expert opinion.
efficacy of reversal of symptoms by replacement  Two groups (ISA and Endocrine Society)
testosterone treatment, but it is under-powered for have attempted evidence-based
guidelines, but both depend significantly on
judging cardiovascular disease and prostate safety
expert opinion.
issues. An interventional arm of the European Male
 Highest quality, evidence-based guidelines
Aging Study (EMAS) will provide valuable add- require appropriately powered, well-controlled
itional data as the age groups are wider and the (ideally double-blinded, placebo-controlled)
populations differ. data that has been validated by confirmatory
It is also obvious to the present authors that the studies.
lack of specificity of the signs and symptoms of low  Evidence-based guidelines for hypogonadism are
serum testosterone, the uncertainty in the appropriate considered to be of low quality.
tests to identify hypogonadism, the lack of a clear  Guidelines based on expert opinion depend on
threshold of serum testosterone levels for clinical the selection of the expert panel and are subject
phenotype, and the great impact of co-morbid condi- to uncertain validity.
tions on the prevalence rate of androgen deficiency  Guidelines on hypogonadism share some
common conclusions but differ in diagnostic
has led to uncertainty whether low serum testosterone
criteria, treatment indications and monitoring
is a cause of a disorder or a manifestation associated strategies.
with many disorders. This is particularly true in  Guidelines are living documents and require
androgen deficiency in older men. revision as new data of improved quality become
Furthermore, the recommendations on serum tes- available.
tosterone goals for treatment and the type and

418
Chapter 18: Guidelines for diagnosis and treatment

18.11 References Zhang A, Choong K, Lakshman


KM, Mazer NA, Miciek R, Krasnoff
bone metabolism and serum lipid
profile in middle-aged men: a
Abrahamsson U, Arver S, Damber JE, J, Elmi A, Knapp PE, Brooks B, meta-analysis. Clin Endocrinol (Oxf )
Ekstrom U, Giwercman A, Lehtihet Appleman E, Aggarwal S, Bhasin G, 63:280293
M, Link K, Nilsson J, Peeker R, Hede-Brierley L, Bhatia A, Collins
Pousette A, Rosen T, Stroberg P, Ly LP, Sartorius G, Hull L, Leung A,
L, LeBrasseur N, Fiore LD, Bhasin S Swerdloff RS, Wang C, Handelsman
Willenheimer R (2010) (2010) Adverse events associated
[Testosterone Deficiency DJ (2010) Accuracy of calculated
with testosterone administration. free testosterone formulae in men.
Management Programme and N Engl J Med 363:109122
Treatment Recommendations.] Clin Endocrinol (Oxf ) 73:382388
Svensk Andrologisk Frening, Basurto L, Zarate A, Gomez R, Vargas McLachlan RI (2010) Certainly more
Stockholm. Available at: www. C, Saucedo R, Galvan R (2008) guidelines than rules. J Clin
svenskandrologi.se/res/Default/ Effect of testosterone therapy on Endocrinol Metab 95:26102613
vardprogram.pdf (accessed lumbar spine and hip mineral
density in elderly men. Aging Male Meier CT, Nguyen V, Handelsman DJ,
Feb 22, 2012)
11:140145 Schindler C, Kushnir MM,
American Society for Reproductive Rockwood AL, Meikle AW, Center
Medicine (2006) Treatment of Behre HM, Kliesch S, Leifke E, Link JR, Eisman JA, Seibel MJ (2008)
androgen deficiency in the aging TM, Nieschlag E (1997) Long-term Endogenous sex hormones and
male. Fertil Steril 86:S236S240 effect of testosterone therapy on incident fracture risk in older men:
bone mineral density in the Dubbo Osteoporosis
Amin S, Zhang Y, Sawin CT, Evans SR,
hypogonadal men. J Clin Endocrinol Epidemiology Study. Arch Intern
Hannan MT, Kiel DP, Wilson PW,
Metab 82:23862390 Med 168:4754
Felson DT (2000) Association of
hypogonadism and estradiol levels Bhasin S, Cunningham GR, Hayes FJ, Merza Z, Blumsohn A, Mah PM,
with bone mineral density in elderly Matsumoto AM, Snyder PJ, Meads DM, McKenna SP, Wylie K,
men from the Framingham study. Swerdloff RS, Montori VM (2010) Eastell R, Wu F, Ross RJ (2006)
Ann Intern Med 133:951963 Testosterone therapy in men with Double-blind placebo-controlled
androgen deficiency syndromes: an study of testosterone patch therapy
Amory JK, Watts NB, Easley KA,
Endocrine Society clinical practice on bone turnover in men with
Sutton PR, Anawalt BD, Matsumoto
guideline. J Clin Endocrinol Metab borderline hypogonadism. Int
AM, Bremner WJ, Tenover JL
95:25362559 J Androl 29:381391
(2004) Exogenous testosterone or
testosterone with finasteride Brand JS, van der Tweel I, Grobbee DE, Morales A, Bella AJ, Chun S, Lee J,
increases bone mineral density in Emmelot-Vonk MH, van der Assimakopoulos P, Bebb R,
older men with low serum Schouw YT (2010) Testosterone, sex Gottesman I, Alarie P, Dugre H,
testosterone. J Clin Endocrinol hormone-binding globulin and the Elliott S (2010) A practical guide to
Metab 89:503510 metabolic syndrome: a systematic diagnosis, management, and
review and meta-analysis of treatment of testosterone deficiency
Anawalt BD (2010) Guidelines for
observational studies. Int for Canadian Physicians. Can Urol
testosterone therapy for men: how
J Epidemiol 40:189207 Assoc J 4:269275
to avoid a mad (t)ea party by getting
personal. J Clin Endocrinol Metab Conway AJ, Handelsman DJ, Lording Morley JE, Charlton E, Patrick P,
95:26142617 DW, Stuckey B, Zajac JD (2000) Kaiser FE, Cadeau P, McCready D,
Andriole GL, Crawford ED, Grubb RL Use, misuse and abuse of Perry HM III (2000) Validation of a
3rd, Buys SS, Chia D, Church TR, androgens. The Endocrine Society screening questionnaire for
Fouad MN, Gelmann EP, Kvale PA, of Australia consensus guidelines androgen deficiency in aging males.
Reding DJ, Weissfeld JL, Yokochi for androgen prescribing. Med Metabolism 49:12391242
LA, OBrien B, Clapp JD, Rathmell J Aust 172:220224
Nair KS, Rizza RA, OBrien P,
JM, Riley TL, Hayes RB, Kramer BS, Heinemann LA, Saad F, Heinemann K, Dhatariya K, Short KR, Nehra A,
Izmirlian G, Miller AB, Pinsky PF, Thai DM (2004) Can results of the Vittone JL, Klee GG, Basu A, Basu
Prorok PC, Gohagan JK, Berg CD Aging Males Symptoms (AMS) R, Cobelli C, Toffolo G, Dalla Man
(2009) Mortality results from a scale predict those of screening C, Tindall DJ, Melton LJ 3rd, Smith
randomized prostate-cancer scales for androgen deficiency? GE, Khosla S, Jensen MD (2006)
screening trial. N Engl J Med Aging Male 7:211218 DHEA in elderly women and
360:13101319 Isidori AM, Giannetta E, Greco EA, DHEA or testosterone in elderly
Basaria S, Coviello AD, Travison TG, Gianfrilli D, Bonifacio V, Isidori A, men. N Engl J Med 355:16471659
Storer TW, Farwell WR, Jette AM, Lenzi A, Fabbri A (2005) Effects of Page ST, Amory JK, Bowman FD,
Eder R, Tennstedt S, Ulloor J, testosterone on body composition, Anawalt BD, Matsumoto AM,

419
Chapter 18: Guidelines for diagnosis and treatment

Bremner WJ, Tenover JL (2005) Snyder PJ, Peachey H, Hannoush P, mood, lean and fat mass, and bone
Exogenous testosterone (T) alone or Berlin JA, Loh L, Holmes JH, mineral density in hypogonadal
with finasteride increases physical Dlewati A, Staley J, Santanna J, men. J Clin Endocrinol Metab
performance, grip strength, and Kapoor SC, Attie MF, Haddad JG Jr, 89:20852098
lean body mass in older men with Strom BL (1999) Effect of
Wang C, Nieschlag E, Swerdloff RS,
low serum T. J Clin Endocrinol testosterone treatment on bone
Behre HM, Hellstrom WJ, Gooren
Metab 90:15021510 mineral density in men over 65
LJ, Kaufman JM, Legros JJ,
Petak SM, Nankin HR, Spark RF, years of age. J Clin Endocrinol
Lunenfeld B, Morales A, Morley JE,
Swerdloff RS, Rodriguez-Rigau LJ Metab 84:19661972
Schulman C, Thompson IM,
(2002) American Association of Srinivas-Shankar U, Roberts SA, Weidner W, Wu FC (2009)
Clinical Endocrinologists Medical Connolly MJ, OConnell MD, Investigation, treatment, and
Guidelines for clinical practice for Adams JE, Oldham JA, Wu FC monitoring of late-onset
the evaluation and treatment of (2010) Effects of testosterone on hypogonadism in males: ISA,
hypogonadism in adult male muscle strength, physical function, ISSAM, EAU, EAA, and ASA
patients 2002 update. Endocr Pract body composition, and quality of recommendations. J Androl 30:19
8:440456 life in intermediate-frail and frail
Wang C, Jackson G, Jones TH,
Rosen RC, Araujo AB, Connor MK, elderly men: a randomized, double-
Matsumoto AM, Nehra A,
Gerstenberger EP, Morgentaler A, blind, placebo-controlled study.
Perelman MA, Swerdloff RS, Traish
Seftel AD, Miner MM, Shabsigh R J Clin Endocrinol Metab 95:639650
A, Zitzmann M, Cunningham G
(2011) The NERI Hypogonadism Svartberg J, Agledahl I, Figenschau Y, (2011) Low testosterone associated
Screener: psychometric validation Sildnes T, Waterloo K, Jorde R with obesity and the metabolic
in male patients and controls. (2008) Testosterone treatment in syndrome contributes to sexual
Clin Endocrinol 74:248256 elderly men with subnormal dysfunction and cardiovascular
Sartorius G, Ly LP, Sikaris K, testosterone levels improves body disease risk in men with type
McLachlan R, Handelsman DJ composition and BMD in the hip. 2 diabetes. Diabetes Care
(2009) Predictive accuracy and Int J Impot Res 20:378387 34:16691675
sources of variability in calculated Tracz MJ, Sideras K, Bolona ER, Wolf AM, Wender RC, Etzioni RB,
free testosterone estimates. Ann Clin Haddad RM, Kennedy CC, Uraga Thompson IM, DAmico AV, Volk
Biochem 46:137143 MV, Caples SM, Erwin PJ, Montori RJ, Brooks DD, Dash C, Guessous I,
Schroder FH, Hugosson J, Roobol MJ, VM (2006) Testosterone use in men Andrews K, DeSantis C, Smith RA;
Tammela TL, Ciatto S, Nelen V, and its effects on bone health. American Cancer Society Prostate
Kwiatkowski M, Lujan M, Lilja H, A systematic review and meta- Cancer Advisory Committee (2010)
Zappa M, Denis LJ, Recker F, analysis of randomized placebo- American Cancer Society guideline
Berenguer A, Maattanen L, Bangma controlled trials. J Clin Endocrinol for the early detection of prostate
CH, Aus G, Villers A, Rebillard X, Metab 91:20112016 cancer: update 2010. CA Cancer
van der Kwast T, Blijenberg BG, Vermeulen A, Verdonck L, Kaufman J Clin 60:7098
Moss SM, de Koning HJ, Auvinen A JM (1999) A critical evaluation of Wu FC, Tajar A, Beynon JM, Pye SR,
(2009) Screening and prostate- simple methods for the estimation Silman AJ, Finn JD, ONeill TW,
cancer mortality in a randomized of free testosterone in serum. J Clin Bartfai G, Casanueva FF, Forti G,
European study. N Engl J Med Endocrinol Metab 84:36663672 Giwercman A, Han TS, Kula K,
360:13201328 Wang C, Cunningham G, Dobs A, Lean ME, Pendleton N, Punab M,
Smith KW, Feldman HA, McKinlay JB Iranmanesh A, Matsumoto AM, Boonen S, Vanderschueren D,
(2000) Construction and field Snyder PJ, Weber T, Berman N, Labrie F, Huhtaniemi IT (2010)
validation of a self-administered Hull L, Swerdloff RS (2004) Long- Identification of late-onset
screener for testosterone deficiency term testosterone gel (AndroGel) hypogonadism in middle-aged
(hypogonadism) in ageing men. treatment maintains beneficial and elderly men. N Engl J Med
Clin Endocrinol 53:703711 effects on sexual function and 363:123135

420
Chapter
Pathophysiology of estrogen action

19 in men
Vincenzo Rochira, Daniele Santi, and Cesare Carani

19.1 Estrogens in men: historical 19.4.1 Gender identity and sexual


development 421 orientation 428
19.2 Estrogen action on the hypothalamic- 19.4.2 Sexual behavior 429
pituitary unit: regulation of gonadotropin 19.5 Estrogens and the male bone 429
feedback 423 19.5.1 Bone maturation 430
19.3 Estrogens and male reproduction 425 19.5.2 Bone mineral density 431
19.3.1 Estrogen effects on male reproduction 19.6 Estrogens and metabolism 431
in rodents 426 19.6.1 Glucose metabolism 431
19.3.2 Knock-out models of estrogen 19.6.2 Lipid metabolism 432
deficiency 427 19.7 Clinical and therapeutic implications 432
19.3.3 Estrogen effects on male reproduction 19.8 Conclusions 432
in men 427
19.9 Key messages 433
19.3.4 Inappropriate estrogen exposure 427
19.10 References 433
19.4 Estrogens and human male sexuality 428

19.1 Estrogens in men: historical ligands in male tissues, including the reproductive
development system (Greco et al. 1992; Brodie and Inkster 1993),
Adult male sexual and reproductive functions were together with the advancements in testicular paracri-
traditionally considered to be under the control of nology (Saez 1994), all contributed to expand the
both gonadotropins and androgens. Hence, testoster- comprehension of estrogens role in men. Although
one has long been considered the male hormone the concept of testosterone as a prohormone for
(Funk et al. 1930), and the concept of estrogen estrogen in men was old (Zondek 1934), the role of
involvement on male physiology is new. In 1934 estrogens in men continued to be uncertain until the
Zondek, however, first documented estrogen produc- beginning of the nineties, and estrogens were left in
tion in the male by demonstrating the intratesticular the background for a long time in male physiology.
conversion of androgens into estrogens in male stal- The idea that estrogens regulate several functions
lions (Zondek 1934). This mechanism was assumed in men, also including human male reproduction, is
to operate in men as well, but the real demonstration a recent acquisition (Faustini-Fustini et al. 1999;
of estrogen production in the human male was pro- Rochira et al. 2005), that has emerged only in the last
vided only some years later in 1979 (MacDonald et al. 20 years (for review see Rochira et al. 2005), when the
1979). Subsequent observations and studies, such as development of molecular biology allowed detailed
the characterization of the biochemical pathways characterization of estrogen receptors and their func-
(Heard et al. 1955; MacDonald et al. 1979) and the tion (Couse et al. 1997), as well as the genes involved
immunohistochemical demonstration of the estrogen in the synthesis of estrogens (Simpson et al. 1994).

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

421
Chapter 19: Pathophysiology of estrogen action in men

H H
O
H Progesterone

H H
HO 17-hydroxylase
Cholesterol

OH
H

H H
O
17-OH-progesterone

17,20-lyase

O O
CH3

H H Organs:

H H Testis
H H
O Adipose tissue
HO
Aromatase Androstenedione
Estrone Muscle tissue
Brain
Liver
CH3 OH OH Breast
Aromatase Skin
H H
Endothelium

H H
H H Gastric parietal cells
O
HO
Testosterone
Estradiol
Fig. 19.1 Estradiol biosynthesis in men.

A further step was taken when animal models for In males, estrogens derive from circulating andro-
congenital estrogen deficiency were generated (Korach gens and are mainly produced by the testes, adipose
1994; Couse and Korach 1999). All the studies men- and muscle tissue (Fig. 19.1). The key step for estrogen
tioned above, together with the discovery of mutations biosynthesis is the aromatization of C19 androgens,
in the estrogen receptor alpha (ERa) (Smith et al. which are responsible for testosterone and androstene-
1994), and/or in the aromatase enzyme in humans dione conversion into estradiol and estrone, respectively
(Morishima et al. 1995; Carani et al. 1997) definitively (Fig. 19.1). This conversion is regulated by the aromatase
changed some classical standpoints in endocrinology, enzyme, a protein that belongs to the family of P450
providing evidence that estrogens also exert a wide mono-oxygenase enzyme complex (Simpson et al. 1994;
range of biological effects in men and not only in Fig. 19.1). This enzyme is responsible for three consecu-
women (Deroo and Korach 2006). tive hydroxylation reactions, the final result of which is

422
Chapter 19: Pathophysiology of estrogen action in men

Mutation of human ER gene


leads to estrogen-resistance
clinical characterization
Mutation of human aromatase
Development of testicular gene leads to aromatase clinical
paracrinology and local paracrine characterization
Estradiol effects network
on male sexual Estrogen required for
development Biochemical evidence growth arrest in men
of androgen aromatization
into estrogen Estrogens act through a
threshold in male bone

1955 1979 1980 1994 1995


1934 1940 2001 2011
1972 1987 1985 1997 1993 1995

Estradiol production by
Testicular production of germinal cells
estrogen in men
Generation of knock-out
mice models for ER,
Estradiol production Immunohistochemical ER and aromatase
within studies enzyme
stallion testes
ER, ER and CYP19 human
gene cloning
Fig. 19.2 Estrogens in men: milestones.

the aromatization of the A-ring of androgens (Simpson testosterone is required for normal functioning of the
et al. 1994; Gruber et al. 2002; Fig. 19.1). hypothalamic-pituitary testicular axis (Faustini-Fustini
Circulating estrogens are reversibly bound, mainly et al. 1999; Simoni et al. 1999). Accordingly, serum
to SHBG and, to a lesser degree, to albumin. Estrogen gonadotropins are high in all adult patients with
actions are mediated by binding to specific nuclear aromatase deficiency, in the presence of normal to
estrogen receptors (ERs), which are ligand-inducible increased serum testosterone (Rochira and Carani
transcription factors regulating the expression of target 2009; Table 19.1 and Table 19.2), thus implying that
genes (Gruber et al. 2002; Deroo and Korach 2006). Two estrogens are important for regulating the amount
subtypes of nuclear ERs have been described (Gruber of circulating gonadotropins in men. A detailed study
et al. 2002): ERa and the more recently discovered ERb. of the effects of different doses of transdermal estra-
Other than the classical pathways based on tran- diol on pituitary function in a man with congenital
scriptional mechanisms, a non-genomic action enrol- aromatase deficiency demonstrated that estrogens
ling cell-surface receptors of estrogens has been might control not only basal secretion of gonadotro-
recently documented (Gruber et al. 2002). pins, but also their responsiveness to GnRH adminis-
Milestones of knowledge referring to estrogen tration. In this study, estrogen administration to a
physiology in men (for review see Rochira et al. male patient with aromatase deficiency decreased
2005) are summarized in Fig. 19.2. basal and GnRH-stimulated secretion of LH, FSH
and a-subunit, in a dose-dependent manner (Rochira
19.2 Estrogen action on the et al. 2002a). Furthermore, in men without congenital
hypothalamic-pituitary unit: regulation defects of estrogen function, the pharmacological
inhibition of estrogen also results in a significant
of gonadotropin feedback increase of both LH and FSH (Taxel et al. 2001;
Animal and human models of congenital estrogen De Ronde and de Jong 2011; Saylam et al. 2011). All
deficiency provide evidence that aromatization of these data support an inhibitory effect of estrogens

423
Chapter 19: Pathophysiology of estrogen action in men

Table 19.1 Phenotype of animal models of estrogen resistance and aromatase deficiency

aERKO bERKO Estrogen ArKO mice Aromatase deficiency


mice resistance in in men
men (ERa)
Testis Germ cells loss; Normal Normal Normal at 14 wk Normal or increased
enlarged volume
seminiferous tubule (2025 ml)
Germ cells Normal Not Not described Disruption of Germ cell arrest at
development described spermatogenesis spermatocyte level
of germ cells when at 1 year of age Or
transplanted in the Complete germ cell
wild type depletion
Sperm Reduced number; Normal Normal sperm Reduced sperm From
characteristics motility and sperm count count and oligoasthenozoospermia
fertilizing capacity count (25  106/ml) decreased to severe
Reduced viability at asthenospermia
viability (18%) 8 months of age
Fertility Infertile Fertile Fertile? Infertile at 1 year Infertile
Hormonal LH " LH ! LH " LH " LH ! "
Pattern FSH ! FSH ! FSH " FSH ! FSH "
T" T! T! T" T!"
E 2" E2 ! E2 " E2 Undetectable E2 Undetectable
Abbreviations: T, testosterone; E2, estradiol.
Symbols: " increase; ! unchanged.

on gonadotropin secretion at the pituitary level deficiency (Pitteloud et al. 2008a), as well as in aroma-
(Faustini-Fustini et al. 1999; Simoni et al. 1999; Fig. tase-deficient men (Rochira et al. 2006a).
19.3) that operates from early to mid-puberty (Mauras Estrogens are also able to modulate FSH secretion
et al. 2000; Wickman and Dunkel 2001) and that in men (Rochira et al. 2001; 2005). In normal men
persists even during adulthood and advanced age with pharmacologically induced sex steroid depriv-
(Taxel et al. 2001). The hypothalamic-pituitary gonadal ation, estradiol, but not testosterone, is able to restore
axis, however, also responds to androgens, since the normal FSH serum levels (Pitteloud et al. 2008b). Due
administration of DHT is able to decrease LH and FSH to the concomitant impairment of the patients
partially with a concomitant reduction in serum testos- spermatogenesis, complete normalization of serum
terone and estradiol (Idan et al. 2010). FSH was not achieved in all aromatase-deficient
More recently, estrogen action at hypothalamic men during estradiol treatment, even in the presence
level has also been documented (Raven et al. 2006; of physiological levels of circulating estradiol
Rochira et al. 2006a), and the main influence is exerted (Rochira and Carani 2009); only supraphysiological
by circulating estrogens, rather than by those locally levels of estrogens being necessary to obtain FSH
produced (Raven et al. 2006; Rochira et al. 2006a; normalization (Carani et al. 1997; Rochira et al.
Fig. 19.3). The administration of anastrozole, a potent 2000).
aromatase inhibitor, to men with IHH resulted, in fact, Other than the gonadal axis, even growth hor-
in a decrease of GnRH pulse frequency at the hypo- mone, prolactin and thyroid-stimulating hormone
thalamic level and in a blunted pituitary responsiveness are partially modulated by estrogens in men, as
to GnRH (Hayes et al. 2000). Furthermore, estradiol documented by their abnormal secretion in men
administration lowers LH pulse frequency in normal with aromatase deficiency (Rochira et al. 2002a;
subjects with pharmacologically induced estrogen Rochira and Carani 2009).

424
Chapter 19: Pathophysiology of estrogen action in men

Table 19.2 Summary of both well-established and supposed estrogen actions on male reproductive system

Function Animals Humans


Spermatogenesis Well- Fluid reabsorption in the efferent Abnormal development of male
established ductules (ERa) reproductive structures after exposure
Sperm concentration (ERa) to estrogen excess
Abnormal development of male
reproductive structures after exposure to
estrogen excess
Supposed Growth control of germ cells Control of spermatogenesis and sperm
proliferation during fetal life (ERb) maturation
Germ cell differentiation
Inhibition of germ cell apoptosis (ERb)
Control of cell adhesion (particularly on
Sertoli cells)
Gonadotropin Well- Inhibition of gonadotropin secretion at Inhibition of gonadotropin secretion at
secretion established pituitary level both pituitary and hypothalamic level
Supposed Inhibition of gonadotropin secretion at
hypothalamic level
Sexual behavior Well- Promotion of mating copulative No effects on gender identity and
established behavior sexual orientation
Supposed Determinant for partner preference Possible positive role on male sexual
behavior
Bone physiology Well- Bone mineral density maintenance Epiphyseal closure and growth arrest
established Acquisition of normal skeletal
proportions
Acquisition of peak bone mass
Maintenance of bone mass
Supposed Anabolic effect on cortical bone
Action on bone by means of a threshold
(between 12 and 20 pg/ml of serum
estradiol)
Glucose and lipid Well- Modulation of insulin sensitivity and Control of insulin sensitivity
metabolism established glucose tolerance Control of HDL-C production
Modulation of fat storage and adiposity
Supposed Regulation of lipid metabolism Control of glucose tolerance
Regulation of total and LDL cholesterol
Regulation of fat distribution

In conclusion, the control of gonadotropin are expressed and functionally active throughout the
feedback exerted by sex steroids is mainly related male reproductive tract (Couse and Korach 1999;
to estrogens, particularly to the amount circulating ODonnell et al. 2001). Thus, it is now clear that
(Fig. 19.3). the presence of estrogens and ERs in the male repro-
ductive system ensures a physiological function of
19.3 Estrogens and male estrogens on male reproduction, and that both estro-
gen deficiency and excessive estrogen exposure
reproduction may have deleterious reproductive effects in men
In rodents and in men, immunocytochemical studies (Sharpe 1998; Rochira et al. 2001; 2005; Rochira
have revealed that ERs and the aromatase enzyme and Carani 2009).

425
Chapter 19: Pathophysiology of estrogen action in men

in situ Fig. 19.3 Estrogens role at


T DHT E2 hypothalamic and pituitary level.
() GnRH neurons ()
()
in situ
T E2
P450 arom

in situ
T E2 () LH () E2
P450 arom
Circulating sex steroids

Testosterone (T)
Estradiol (E2) E2

19.3.1 Estrogen effects on male and Hess 2010). The lack of estradiol may
promote apoptosis, with a resulting failure in
reproduction in rodents elongated spermatid differentiation (Pentakainen
The site of ERs and aromatase expression varies et al. 2000).
widely during development in rodents, and they are Estrogen-deficient knock-out mice are useful
expressed at a very early stage (Rochira et al. 2005; models to investigate estrogen action in rodents
2009). ERa is abundant in efferent ductules from fetal (Couse and Korach 1999; ODonnell et al. 2001).
life to adulthood, implying a crucial role in male At present, four different lines of estrogen-deficient
reproduction (Hess et al. 2011); while ERb is mainly knock-out mice have been generated (Table 19.1):
expressed during fetal life, suggesting a major role in (1) ERa knock-out (aERKO) mice with disrupted
the development of male reproductive structures until ERa gene; (2) the bERKO mice, with an inactiva-
birth (ODonnell et al. 2001; Rochira et al. 2009). ted ERb; and (3) the abERKO mice with non-
Accordingly, ERb is probably the main target functioning ERa and ERb (Couse and Korach
accounting for the effects of supraphysiological estro- 1999). The aERKO, bERKO and abERKO mice
gen levels on abnormal development of reproductive provide helpful information on the loss of ER
structures during fetal life (Toppari and function (estrogen resistance). The knock-out of
Skakkebaek 1998; ODonnell et al. 2001; Rochira aromatase gene leads to aromatase knock-out
et al. 2009). In the adult male reproductive system (ArKO) mice, an experimental animal model useful
of rodents, ERa is mainly expressed in the proximal for investigating the congenital lack of both circu-
(rete testis, efferent ductules, proximal epididymis), lating and locally produced estrogens since birth
rather than in the distal (corpus and cauda of (Korach 1994; Couse and Korach 1999; ODonnell
the epididymis, vas deferens) reproductive ducts et al. 2001). Estrogen-resistant knock-out mice
(ODonnell et al. 2001; Rochira et al. 2009). This have high levels of circulating estrogens with also
peculiar distribution accounts for several important the non-genomic pathway still functioning, while
estrogen actions in the proximal ducts; especially the aromatase-deficient mice have no circulating estro-
efferent ductules (Hess et al. 2011). gens and no active estrogen pathway (ODonnell
Estradiol may serve as a survival factor for et al. 2001). The reproductive phenotype of
round spermatids and to maintain spermatogenesis estrogen-deficient knock-out mice is summarized
and promote normal sperm maturation (Carreau in Table 19.1.

426
Chapter 19: Pathophysiology of estrogen action in men

19.3.2 Knock-out models of but also in mature human spermatozoa (Pentakainen


et al. 2000; Aquila et al. 2004). Thus, human sperm-
estrogen deficiency atozoa are a site of estrogen biosynthesis and they
Male aERKO mice are infertile since the seminiferous should be considered a mobile endocrine unit produ-
epithelium is atrophic and degenerated, and both cing and responding to estrogens (Rochira et al. 2009;
tubules and rete testis are dilated (Hess et al. 2011). Carreau and Hess 2010).
The lack of estrogen-dependent fluid reabsorption Data from human subjects with congenital estro-
leads to increased fluid back-pressure, resulting in a gen deficiency document that all the eight affected
retroactive progressive swelling of the seminiferous men had an impairment of spermatogenesis, ranging
tubules caused by the marked dilation of the tubules from severe oligozoospermia and oligoasthenosper-
(Couse and Korach 1999; Hess et al. 2011; Table 19.1). mia to asthenospermia (Rochira et al. 2005; Zirilli
In aERKO mice, serum LH is increased, inducing et al. 2008; Rochira and Carani 2009). The variable
Leydig cell hyperplasia and increased serum testoster- degree of fertility impairment in men with congenital
one while FSH is normal (Korach 1994; Couse and deficiency of estrogen action or synthesis does not
Korach 1999; Table 19.1). allow any conclusions to be drawn, and whether these
Unlike aERKO mice, male ArKO mice are ini- reproductive features are the consequence of a con-
tially fully fertile, but fertility decreases with advan- genital lack of estrogen or they are only epiphenom-
cing age due to complete arrest of spermatogenesis at ena remains unsolved. Outside the context of estrogen
the level of early spermatids (Couse and Korach deficiency, however, administration of aromatase
1999). The early arrest of spermatogenesis suggests a inhibitors to infertile men with impaired testoster-
failure in germ cell differentiation, as a consequence one-to-estradiol ratio resulted in an improvement of
of estrogen lack in the testicular environment (Luconi their fertility rate (Raman and Schlegel 2002; Saylam
et al. 2002), leading to a less severe degree of infer- et al. 2011), suggesting the importance of estrogens
tility than in aERKO mice (Couse and Korach 1999). in male reproduction. Finally, the role of estrogen in
Conversely, bERKO mice are fully fertile, also in controlling sperm production and quality has been
adulthood (Couse and Korach 1999; Table 19.1). recently confirmed by the association of polymor-
Gonadotropins are increased in ArKO mice due to phisms of estrogen-related genes with both sperm
the lack of an estrogen-inhibitory effect at the central concentration and motility, but not with sperm
level (see above) (Table 19.1). morphology (Lee et al. 2011).
In conclusion, a functional ERa, but not ERb, is
needed for the development and maintenance of
normal fertility in male mice (Rochira et al. 2009). 19.3.4 Inappropriate estrogen exposure
Many studies in animals suggest that inappropriate
19.3.3 Estrogen effects on male exposure to estrogens in utero and during the neo-
natal period impairs testicular descent, the hypothal-
reproduction in men amic-pituitary-gonadal axis and testicular function;
Aromatase enzyme, ERa and ERb are widely in particular prenatal exposure to diethylstilbestrol
expressed throughout the male reproductive system, (DES) results in abnormal development of male
even in men, and they are also found in human sperm reproductive structures (Sharpe 1998; Toppari and
(Rochira et al. 2005; 2009; Carreau and Hess 2010). Skakkebaek 1998).
Accordingly, ERs are present in the human sperm In men, increased estrogen exposure has been
membrane (Luconi et al. 1999), where they modulate supposed to impair fertility (Sharpe 1998; Toppari
a rapid non-genomic action. Thus, human ejaculated and Skakkebaek 1998), on the basis of the progressive
spermatozoa are responsive to estrogens throughout decline in sperm count and the corresponding
their run from the testes to the urethra, since they also increase of environmental estrogens in some western
express ERa and ERb, and it seems that estrogens countries. In the past, the inappropriate clinical use of
modulate sperm maturation (Luconi et al. 1999; DES in pregnant women was associated with a rise in
Carreau and Hess 2010). the incidence of genital malformations at birth such
In the human testis both somatic and germ cells as: epididymal cysts, meatal stenosis, hypospadias,
express aromatase, not only in immature germ cells, cryptorchidism and microphallus. In addition, semen

427
Chapter 19: Pathophysiology of estrogen action in men

quality of men exposed to DES in utero resulted in structures between men and women (Drner 1988;
greater impairment than in unexposed controls, but LeVay 1991). Prenatal hormonal exposure is classic-
without clear indication of subfertility or clinical ally considered to be involved in determining sexual
infertility (Wilcox et al. 1995). orientation on the basis of some differences in hypo-
In conclusion, animal studies strongly support the thalamic structures found between heterosexual and
idea that exposure to estrogen excess affects the devel- homosexual men (LeVay 1991; Garcia-Falgueras and
opment and function of reproductive male organs, Swaab 2010). This hypothesis is supported by the
but the real impact of estrogen overexposure on concept that sexual differentiation of the brain takes
human male fertility remains to be established. place in parallel with the peak of testosterone secre-
However, the exact mode and site of action of tion from the testis and the corresponding increase in
estrogens in the male reproductive system remain serum estradiol during fetal life (Gorski 1991; Roselli
only partially known, and further research is required et al. 2009; Garcia-Falgueras and Swaab 2010).
to improve knowledge of this issue. According to this hypothesis, the intrinsic pattern of
mammalian brain development is female, and estro-
19.4 Estrogens and human gen is required for the development of a male brain
(Drner 1988; Garcia-Falgueras and Swaab 2010);
male sexuality thus emphasizing the role of estrogen aromatization
19.4.1 Gender identity and sexual locally in the brain (Roselli et al. 2009). Permanent
changes in the organization of certain neural circuits,
orientation a prerequisite for sex-specific regulation of reproduc-
Sex steroids, mainly testosterone, modulate adult tive and sexual behavior, probably also occur under
male sexual behavior in mammals (Robbins 1996). the effects of estrogen (Pilgrim and Reisert 1992;
In men, sexual behavior is the result of the sum of Roselli et al. 2009; Garcia-Falgueras and Swaab
the effects of cognitive processes, cultural environ- 2010). By considering all the factors mentioned above,
ment, hormonal and genetic prerequisites (Rochira the lack of estrogen action on the developing brain
et al. 2003). The role of estrogen in male sexual in males was believed to be strictly related to both
behavior has been poorly investigated, and knowledge dimorphism of hypothalamic structures, and the
derives mainly from studies performed on animals or future development of sexual orientation (Gorski
from rare models of human estrogen deficiency. 1991; LeVay 1991; Roselli et al. 2009; Garcia-
Testosterone aromatization to estradiol in the Falgueras and Swaab 2010), even though most of the
brain was considered the key step in the establishment data came from studies performed in rodents (Pilgrim
of a male brain and in determining sexual dimorph- and Reisert 1992; Roselli et al. 2009; Garcia-Falgueras
ism of the central nervous system in non-primate and Swaab 2010). Recently, the role for local hypo-
mammals (Gorski 1991; Pilgrim and Reisert 1992; thalamic aromatase activity and expression in partner
Garcia-Falgueras and Swaab 2010). According to preference has been confirmed in rams (Roselli et al.
Drners hypothesis (Drner 1988), prenatal and 2004). In this study, the preferences of sexual partner
perinatal brain exposure to estrogens may be respon- were associated with both the volume of the ovine
sible for the establishment of a male brain (Roselli sexually dimorphic nucleus and different patterns
et al. 2009); an event occurring only in the male brain, of aromatase expression, providing the first demon-
and not in the female one. Accordingly, ovaries stration that differences in aromatase expression
release very small amounts of estrogen that are also within the brain are related to partner preferences
soon inactivated in rodents (Rochira et al. 2005; and are involved in the determination of adult sexual
Roselli et al. 2009), while males produce a greater behavior (Roselli et al. 2004).
amount of androgens that are converted into estro- Aromatase deficiency in men is an interesting
gen. Thus, circulating estrogens are greater in males model to study the role of estradiol in human male
than in females during fetal life, which accounts for sexual behavior by considering the lack of aromatase
the sexual dimorphism of hypothalamic structures in activity from fetal life through adulthood (Rochira
rodents (Roselli et al. 2009; 2011). et al. 2005; 2009; Rochira and Carani 2009). All men
The same mechanism also seems to be involved with aromatase deficiency had a male gender identity
for the establishment of differences in hypothalamic and heterosexual orientation (Morishima et al. 1995;

428
Chapter 19: Pathophysiology of estrogen action in men

Carani et al. 1999; 2005; Herrmann et al. 2002; receptor activation alone is not sufficient for fully
Rochira et al. 2005; 2009; Maffei et al. 2007; normal sexual behavior in rodents (Rochira et al.
Lanfranco et al. 2008; Rochira and Carani 2009). 2005; 2009).
The fact that congenital aromatase deficiency does Much less is known about the role of estrogens in
not affect psychosexual orientation and gender iden- sexual behavior in the human male, since the propor-
tity in humans allows the hypothesis that, in contrast tion of testosterone effects on male sexual behavior
to animals, psychological and social factors may be that may be ascribed to its conversion into estradiol is
the most relevant determinants of gender role behav- still not known. A detailed sexual investigation of
ior in men, with hormones probably having a minor aromatase-deficient men documented an increase of
role (Rochira et al. 2005; Rochira and Carani 2009; all the parameters of sexual activity during estrogen
Roselli et al. 2011). treatment (Carani et al. 1999; 2005), but the best
In conclusion, aromatase has a key role in the outcome in terms of sexual behavior was obtained
control of male reproductive behavior, especially in only when a concomitant normalization of both
animals (rodents and rams), where it induces organ- serum testosterone and estradiol was reached (Carani
izational effects on the developing brain during fetal et al. 2005), supporting the concept that both andro-
life (Wright et al. 2010). Local estrogen production gens and estrogens are required for normal sexual
within the brain and exposure to circulating estrogens behavior in men. Outside the context of congenital
are responsible for testosterone effects on sexual dif- lack of estrogens, it is difficult to obtain conclusive
ferentiation. Differences exist among species, how- information on the role of estrogen on male sexual
ever, and explain the essential role of aromatization behavior because of inadequacy of studies and con-
in rodents (Roselli et al. 2011; Wright et al. 2010) and flicting results in the literature.
its poor effect in humans (Carani et al. 1999; 2005; Finally, estrogen receptors and the aromatase
Rochira et al. 2005; Rochira and Carani 2009). enzyme have been identified in the penile tissue of a
large number of species, including humans (Crescioli
et al. 2003; Dietrich et al. 2004; Goyal et al. 2007),
19.4.2 Sexual behavior suggesting direct estrogenic activity within the penis.
In men, adult male sexual behavior is partially At present, what we know about estrogen action
dependent on testosterone, the main hormone within the penis derives from the observation that
involved in male sexuality (Funk et al. 1930; Granata exposure of male offspring to estrogen-like endocrine
et al. 1997; Rochira et al. 2003; see also Chapter 5). disruptors in utero induces micropenis and hypospa-
Accordingly, testosterone deficiency frequently pro- dias (Toppari et al. 1996) and that, in animals, penile
duces loss of libido and ED (Granata et al. 1997; development and function is estrogen dependent
Rochira et al. 2003). At the same time, TRT increases (Mowa et al. 2006). Estrogens seem to be involved
sexual interest and improves sexual behavior (Robbins in penile post-orgasm flaccidity (Vignozzi et al. 2004),
1996; Rochira et al. 2003). in penile growth and in erection (Crescioli et al. 2003;
In experimental animals, the knock-out of estro- Greco et al. 2006).
gen pathways or a condition of pharmacologically
induced estrogen deficiency results in severe impair-
ment of sexual behavior (Rochira et al. 2005; 2009). 19.5 Estrogens and the male bone
Accordingly, ArKO mice (Honda et al. 1998), Sex steroids, both estrogens and androgens, act on
abERKO male mice and aERKO mice (Couse and bone tissue and influence bone maturation and main-
Korach 1999) all exhibit a significant reduction in tenance. The old concept that estrogens are impli-
mounting frequency and a significant prolonged cated in the regulation of bone metabolism in
latency to mount when compared with wild-type women while androgens regulate the homeostasis of
animals (Couse and Korach 1999; ODonnell et al. the skeleton in men is outdated.
2001). To the contrary, bERKO mice did not show The important role of estrogen in bone metabol-
any defect in any components of sexual behavior, ism in men has been characterized in the last 15 years
including ejaculation (Couse and Korach 1999). by means of the description of rare case reports of
These findings suggest that ERa is required for estrogen-deficient men and by several epidemio-
mounting behavior in male mice and that androgen logical studies (Rochira et al. 2006b; Khosla 2010).

429
Chapter 19: Pathophysiology of estrogen action in men

Estradiol
levels (pg/ml)

Mature bone

20

Fast linear growth with


subsequent closure of epiphyseal
cartilages Bone maturation
and growth arrest and
progressive
epiphyseal closure

12

Continuous progressive
and slow linear growth Cartilage growth

Fig. 19.4 Serum estradiol threshold for epiphyseal closure.

The relative contribution of androgens versus estro- weight, height and pubertal stage (Klein et al. 1996).
gens in the regulation of the male skeleton, however, Besides, estrogens have a biphasic effect on growth
is complex and partially unclear, but some estrogen plates since low doses of estradiol stimulate ulnar
actions on male bone, such as the acceleration of growth in boys, while higher doses lead to an inhib-
growth arrest are now well defined (see also Chapter 8). ition of this process (Cutler 1997).
Nowadays, we know from aromatase-deficient
men that estrogens are necessary in men for the com-
19.5.1 Bone maturation pletion of bone maturation, since they control epiphy-
Pubertal growth in humans was traditionally con- seal closure and growth arrest (Carani et al. 1997;
sidered as an androgen-dependent process since it Herrmann et al. 2002; Maffei et al. 2004). Conversely,
progresses in parallel with the rise in serum testoster- androgens alone are not sufficient to promote normal
one. However, several studies have recently shown skeletal development during puberty (Faustini-Fustini
that part of the actions of androgens at puberty et al. 1999; Rochira et al. 2002b; Zirilli et al. 2008).
(induction of the growth spurt, bone maturation By considering the eight aromatase-deficient men
and ossication of the epiphyseal cartilages) is clearly described to date (Rochira and Carani 2009), a serum
mediated by estrogens in the human male (Zirilli estradiol threshold exists above which bone maturation
et al. 2008). Accordingly, current evidence indicates occurs, and comprises between 12 pg/ml and 20 pg/ml
that estrogens stimulate skeletal maturation in males (Lanfranco et al. 2008; Fig. 19.4). Furthermore, the
as well as in females during many steps of skeletal human model of male aromatase deficiency provides
development (Rochira et al. 2002b; Zirilli et al. 2008). evidence on the pivotal role of estrogens in establishing
Estrogen serum levels increase simultaneously normal skeletal proportions. Accordingly, the coexist-
with testosterone levels throughout puberty, and cor- ence of continuing linear growth of long bones and
relate directly with chronological age, skeletal age, eunuchoid body habitus in men affected by aromatase

430
Chapter 19: Pathophysiology of estrogen action in men

deficiency highlights the concept that estrogens, studies on fracture risk in older men (Mellstrm
rather than androgens, are needed for harmonic et al. 2008; Fig. 19.4), and recently Khosla et al. stated
skeletal growth (Zirilli et al. 2008; Rochira and Carani that a serum estradiol level above 25 pg/ml is certainly
2009). As a consequence, in hypogonadal men the protective for bone in men (Khosla et al. 2011;
disproportional growth of long bones is mainly due Fig. 19.4).
to relative estrogen deficiency (Faustini-Fustini et al. In general, however, the estrogen regulation of
1999; Maffei et al. 2004; Zirilli et al. 2008; Rochira and bone remodeling is due to: (1) inhibition of activation
Carani 2009), as also confirmed by normal skeleton of bone remodeling and of initiation of new basic
proportions in subjects affected by CAIS (Zachmann multicellular units; (2) inhibition of the differen-
et al. 1986). tiation and promoting the apoptosis of osteoclasts,
reducing bone resorption; (3) promotion of the com-
mitment and differentiation of osteoblasts, and pre-
19.5.2 Bone mineral density vention of their apoptosis, maintaining bone
It is now generally accepted that estradiol is the main formation at the cellular level (Khosla et al. 2011).
sex steroid required for bone homeostasis in men For all these reasons, bone loss in man is mainly
(Rochira et al. 2006b). The lack of DHT efficacy in related to relative estrogen deficiency (Rochira et al.
increasing BMD in patients with benign prostate 2006b; Khosla 2010; Khosla et al. 2011).
hyperplasia confirms the minor role of androgen on Even though BMD is strongly estrogen dependent,
bone (Idan et al. 2010). The reduction of lumbar direct action on bone is also exerted in men by andro-
BMD by 1.5% in these patients was the result of the gens, particularly at puberty, probably accounting for
net compensation between the poor anabolic effects sexual dimorphism of the bone mass (Rochira et al.
of high circulating DHT and the prevalent negative 2006b; Vandenput and Ohlsson 2010). A mild to
effect on BMD due to the reduction of testosterone severe reduction of circulating testosterone in men,
and estradiol levels; the latter caused by the negative due to hypogonadism and/or aging, could be respon-
feedback of DHT on gonadotropin secretion (Idan sible for bone loss directly or indirectly through a
et al. 2010). In a recent work, Khosla et al. (2011) concomitant decrease of circulating estradiol. This,
stressed the concept that declining bioavailable estro- with individual differences, may be a determinant
gen levels also made a substantial contribution to age- for the severity of bone clinical manifestations
related bone loss in men, but remarked that cortical (Rochira et al. 2006b).
bone and trabecular bone may respond differently to In conclusion, estrogens seem to be the most
estrogen and to estrogen deficiency. important sex steroid involved in the nal phases of
The relevance of estradiol to bone health derives skeletal maturation and mineralization, even if the
mostly from the evidence that aromatase-deficient exact mode and site of action of estrogens in bone
men have a less mineralized skeleton (Zirilli et al. and in epiphyseal cartilages are not yet completely
2008), and from several studies showing that serum known (Zirilli et al. 2008).
estradiol levels directly correlate with BMD in men
(Khosla 2010). Furthermore, estradiol treatment is
effective in increasing BMD in men with aromatase 19.6 Estrogens and metabolism
deciency, working in a dose-dependent fashion
(Rochira et al. 2000; Zirilli et al. 2008; Rochira and 19.6.1 Glucose metabolism
Carani 2009). Estradiol seems to be protective for The role of estrogen in glucose and insulin metabol-
bone, but only when serum levels are above a thresh- ism in men is difficult to establish since separating
old (Mellstrm et al. 2008); the precise threshold the actions in-vivo of androgen per se from those of
value, however, remains to be settled. In cohort stud- estrogen per se remains challenging. In estrogen-
ies this threshold has been set at around 1220 pg/ml deficient men, insulin resistance and fasting glucose
(Vandenput and Ohlsson 2010), and by studying the are increased and improve during estrogen treatment
effects of estrogen treatment in aromatase-deficient (Rochira et al. 2002b; 2007; Maffei et al. 2004). The
men (Lanfranco et al. 2008), this threshold has been same metabolic pattern is present in knock-out mice
more precisely determined as being around 16 pg/ml models of estrogen deficiency (Couse and Korach 1999).
(Fig. 19.4). This value has also been confirmed by Severe impairment of the estrogen-to-testosterone ratio

431
Chapter 19: Pathophysiology of estrogen action in men

(increased androgens and decreased estrogens) seems when it is important to avoid overtreatment, as in the
to represent a condition for the development of insu- case of testosterone treatment during puberty.
lin resistance in men (Maffei et al. 2004; Rochira et al. Accordingly, the amount of circulating estrogen is
2007), not only in men with congenital estrogen defi- crucial for the closure of bone epiphyses, the achieve-
ciency (Mauvais-Jarvis 2011). ment of final stature and for obtaining both adequate
peak bone mass and skeletal proportions. Thus,
serum estradiol could be a good surrogate for moni-
19.6.2 Lipid metabolism toring testosterone treatment, especially at puberty.
Congenital estrogen deficiency is associated with an Estrogen replacement treatment in aromatase-
altered lipid profile (Faustini-Fustini et al. 1999; deficient men should be started as soon as the diag-
Rochira et al. 2002b), mainly characterized by nosis has been reached, taking into account that
higher total cholesterol and triglyceride serum levels, high doses of estrogen in adult men with aromatase
higher LDL-C and very low HDL-C (Zirilli et al. 2008; deficiency leads to a rapid completion of skeletal
Rochira and Carani 2009). It is noteworthy that maturation within six to nine months, through rapid
HDL-C is inversely linked to serum testosterone; bone elongation and an increase in height followed by
in fact, supraphysiological doses of testosterone quick epiphyseal closure and growth arrest (Rochira
decreased HDL-C in aromatase-deficient men (Maffei et al. 2009). Once epiphyseal closure has been
et al. 2004; Rochira et al. 2007). Conversely, estradiol achieved, estrogen treatment should be continued
treatment determined a moderate increase of HDL-C lifelong, with the main goal of preventing bone loss
together with a small reduction of triglycerides, total and to reduce risk of cardiovascular disease by redu-
cholesterol and LDL-C in aromatase-deficient sub- cing the dose of estradiol in order to maintain serum
jects (Carani et al. 1997; Herrmann et al. 2002; estradiol within the normal range for adult men
Rochira et al. 2002b; Maffei et al. 2004). Overall, the (Rochira and Carani 2009).
effects of estrogen on lipid metabolism resemble those It remains to be established if estrogen in men is
exerted in females (Deroo and Korach 2006). a good target for improving or modulating male
fertility, since conflicting results are available in the
19.7 Clinical and therapeutic area of aromatase inhibitors used for the treatment of
male infertility (Raman and Schlegel 2002; Rochira
implications et al. 2009; Carreau and Hess 2010; Saylam et al.
The knowledge of estrogen pathophysiology in men 2011). The real efficacy of antiestrogens is far from
is of crucial importance for the understanding and being elucidated, and it is a matter of debate whether
clinical management of all conditions of congenital or the increase of sperm density induced by antiestro-
acquired estrogen deficiency in men. gens is actually related to a real improvement of both
Clinical conditions of congenital estrogen defi- sperm fertility and pregnancy rates (Rochira et al.
ciency in men are very rare and include estrogen 2009; Carreau and Hess 2010).
resistance; aromatase deficiency; 17a-hydroxylase-
17,20-lyase deficiency; 17,20-lyase deficiency and cyto-
chrome P450 oxido-reductase deficiency (PORD) 19.8 Conclusions
(Fukami et al. 2009; Rochira and Carani 2009); and Several lines of evidence support the view that estro-
congenital severe hypogonadism. Acquired estrogen gens are required for, and in part mediate, androgen
deficiency is more common in clinical practice and actions on several tissues and organs in men. The
includes transient estrogen deficiency in boys with progress made in the last 20 years in fields of estrogen
delayed puberty, and all the conditions of acquired pathophysiology in men has clarified the importance
severe hypogonadism (Rochira et al. 2006b). of estrogen in men but leaves some issues still unre-
In all these clinical conditions, targeting circulat- solved. In particular, estrogen action on bone and
ing estrogens is of great importance, even in the case gonadotropin secretion is now well characterized
of testosterone replacement treatment, in order to (Table 19.2), and part of the estrogen action on sperm-
ensure adequate effectiveness, such as in the case of atogenesis is now known but needs further evidence.
bone health. Furthermore, estrogen may be useful as a Conversely, the effect of estrogen on glucose and lipid
marker of adequate testosterone treatment, especially metabolism is still controversial (Table 19.2).

432
Chapter 19: Pathophysiology of estrogen action in men

A major area of uncertainty is the possible role of


estrogen in boys before puberty. It is known that low  Estrogens ensure normal skeletal proportions.
levels of circulating estradiol are detected in infancy  Estrogens are necessary for the achievement of
peak bone mass and subsequent maintenance of
when using ultrasensitive assays (Bay et al. 2004), but
bone mineralization.
their significance is not known.  Estrogens exert positive effects on insulin
sensitivity and on levels of HDL cholesterol.
19.9 Key messages
 Congenital estrogen deficiency is characterized
 Circulating estrogens inhibit gonadotropins by the absence of bone maturation, by
by acting at both the hypothalamic and the continuing linear growth, and remains often not
pituitary level. diagnosed until adulthood in men.
 Estrogens probably contribute to modulating  Estrogen replacement treatment is effective on
sperm concentration and motility. bone maturation and mineralization, on the
 Estrogens do not influence gender identity and normal functioning of the
sexual orientation in men. hypothalamic-pituitary-gonadal axis and on
 Estrogens are needed for bone maturation and insulin sensitivity in aromatase-deficient men,
growth arrest in men. but not in estrogen-resistant men.

19.10 References sexual desire in a man with


a novel mutation of aromatase
De Ronde W, de Jong FH (2011)
Aromatase inhibitors in men: effects
Aquila S, Sisci D, Gentile M, Middea E, gene and hypogonadism. and therapeutic options. Reprod Biol
Catalano S, Carpino A, Rago V, Psychoneuroendocrinol 30:413417 Endocrinol 9:93
And S (2004) Estrogen receptor
(ER) alpha and ER beta are both Carreau S, Hess RA (2010) Oestrogens Deroo BJ, Korach KS (2006) Estrogen
expressed in human ejaculated and spermatogenesis. Philos receptors and human disease. J Clin
spermatozoa: evidence of their Trans R Soc Lond B Biol Sci Invest 116:561570
direct interaction with 365:15171535 Dietrich W, Haitel A, Huber JC, Reiter
phospatidylinositol-3-OH kinase/ Couse JF, Korach KS (1999) Estrogen WJ (2004) Expression of estrogen
akt pathway. J Clin Endocrinol receptor null mice: what have we receptors in human corpus
Metab 89:14431451 learned and where will they lead us? cavernosum and male urethra.
Bay K, Andersson AM, Skakkebaek NE Endocrine Rev 20:358417 J Histochem Cytochem 52:355360
(2004) Estradiol levels in Couse JF, Lindzey J, Grandien K, Drner G (1988) Neuroendocrine
prepubertal boys and girls Gustafsson JA, Korach KS (1997) response to estrogen and brain
analytical challenges. Int J Androl Tissue distribution and quantitative differentiation in heterosexuals,
27:266273 analysis of estrogen receptor-a homosexuals, and transsexuals.
Brodie A, Inkster S (1993) Aromatase (ERa) and estrogen receptor-b Arch Sex Behav 17:5775
in the human testis. J Steroid (ERb) messenger ribonucleic acid in
Faustini-Fustini M, Rochira V,
Biochem Molec Biol 44:549555 the wild-type and ERa-knock- Carani C (1999) Oestrogen
Carani C, Qin K, Simoni M, Faustini- out mouse. Endocrinology
deficiency in men: where are we
Fustini M, Serpente S, Boyd J (1997) 138:46134621 today? Eur J Endocrinol 140:111129
Effect of testosterone and estradiol Crescioli C, Maggi M, Vannelli GB,
in a man with aromatase deficiency. Fukami M, Nishimura G, Homma K,
Ferruzzi P, Granchi S, Mancina R, Nagai T, Hanaki K, Uematsu A,
N Engl J Med 337:9195 Muratori M, Forti G, Serio M,
Ishii T, Numakura C, Sawada H,
Carani C, Rochira V, Faustini-Fustini Luconi M (2003) Expression of Nakacho M, Kowase T,
M, Balestrieri A, Granata ARM functional estrogen receptors in Motomura K, Haruna H,
(1999) Role of estrogen in male human fetal male external genitalia. Nakamura M, Ohishi A, Adachi M,
sexual behaviour: insights from the Clin Endocrinol Metab Tajima T, Hasegawa Y, Hasegawa T,
natural model of aromatase 88:18151824 Horikawa R, Fujieda K, Ogata T
deficiency. Clin Endocrinol (Oxf) Cutler GB Jr (1997) The role of (2009) Cytochrome P450
51:517525 estrogen in bone growth and oxidoreductase deficiency:
Carani C, Granata AR, Rochira V, maturation during childhood and identification and characterization
Caffagni G, Aranda C, Antunez P, adolescence, J Steroid Biochem Mol of biallelic mutations and genotype-
Maffei LE (2005) Sex steroid and Biol 61:141144 phenotype correlations in

433
Chapter 19: Pathophysiology of estrogen action in men

35 Japanese patients. J Clin Herrmann BL, Saller B, Janssen OE, the relationship among serum
Endocrinol Metab 94:17231731 Gocke P, Bockish A, Sperling H, estradiol, longitudinal growth and
Mann K, Broecker M (2002) Impact bone mineral density in an adult
Funk C, Harrow B, Lejwa A (1930) The
male hormone. Am J Physiol of estrogen replacement therapy in a man under estrogen replacement
male with congenital aromatase treatment. Bone 43:212218
92:440449
deficiency caused by a novel Lee IW, Kuo PH, Su MT, Kuan LC,
Garcia-Falgueras A, Swaab DF (2010) mutation in the CYP19 gene. J Clin
Hsu CC, Kuo PL (2011)
Sexual hormones and the brain: an Endocrinol Metab 87:54765484 Quantitative trait analysis suggests
essential alliance for sexual identity
Hess RA, Fernandes SA, Gomes GR, polymorphisms of estrogen-related
and sexual orientation. Endocr Dev
Oliveira CA, Lazari MF, Porto CS genes regulate human sperm
17:2235
(2011) Estrogen and its receptors in concentrations and motility. Hum
Gorski RA (1991) Sexual efferent ductules and epidydimis. Reprod 26:15851596
differentiation of the endocrine J Androl 32:600613
brain and its control. In: Motta M LeVay S (1991) A difference in
Honda S, Harada N, Ito S, Takagi Y, hypothalamic structure between
(ed) Brain Endocrinology, 2nd edn. Maeda S (1998) Disruption of
Raven Press, New York, pp 71104 heterosexual and homosexual men.
sexual behavior in male aromatase- Science 253:10341037
Goyal HO, Braden TD, Williams CS, deficient mice lacking exons 1 and 2
Luconi M, Muratori M, Forti G,
Williams J (2007) Role of estrogen of the Cyp19 gene. Biochem Biophys
Baldi E (1999) Identification and
in induction of penile Res Comm 252:445449
characterization of a novel
dysmorphogenesis: a review. Idan A, Griffiths KA, Harwood DT, functional estrogen receptor on
Reproduction 134:199208 Seibel MJ, Turner L, Conway AJ, human sperm membrane which
Granata ARM, Rochira V, Lerchl A, Handelsman DJ (2010) Long-term interferes with progesterone effects.
Marrama P, Carani C (1997) effects of dihydrotestosterone J Clin Endocr Metab 84:16701678
Relationship between sleep-related treatment on prostate growth in
Luconi M, Forti G, Baldi E (2002)
erections and testosterone levels in healthy, middle-aged men without
Genomic and nongenomic effects of
men. J Androl 18:522527 prostate disease: a randomized,
estrogens: molecular mechanisms of
Greco EA, Pili M, Bruzziches R, placebo-controlled trial. Ann Intern
Med 153:621632 action and clinical implications for
Corona G, Spera G, Aversa A (2006) male reproduction. J Steroid Bioch
Testosterone:estradiol ratio changes Khosla S (2010) Update on estrogens Mol Biol 80:369381
associated with long-term taladafil and the skeleton. J Clin Endocrinol
administration: a pilot study. J Sex Metab 95:35693577 MacDonald PC, Madden JD, Brenner
Med 3:716722 PF, Wilson JD, Siiteri PK (1979)
Khosla S, Melton LJ 3rd, Riggs BL Origin of estrogen in normal men
Greco TL, Furlow JD, Duello TM, (2011) The unitary model for and in women with testicular
Gorski J (1992) Immunodetection estrogen deficiency and the feminization. J Clin Endocrinol
of estrogen receptors in fetal pathogenesis of osteoporosis: is a Metab 49:905916
and neonatal male mouse revision needed? J Bone Miner Res
reproductive tracts. Endocrinology 26:441451 Maffei L, Murata Y, Rochira V,
130:421429 Tubert G, Aranda C, Vasquez M,
Klein KO, Martha PO Jr, Blizzard RM, Clyne CD, Davis S, Simpson ER,
Gruber CJ, Tschugguel W, Herbst T, Rogol AD (1996) Carani C (2004) Dysmetabolic
Schneeberger C, Huber J (2002) A longitudinal assessment of syndrome in a man with a novel
Production and actions of hormonal and physical alterations mutation of the aromatase gene:
estrogens. N Engl J Med during normal puberty in boys. II. effects of testosterone, alendronate,
346:340352 Estrogen levels as determined by an and estradiol treatment. J Clin
Hayes FJ, Seminara SB, Decruz S, ultrasensitive bioassay. J Clin Endocrinol Metab 89:6170
Boepple PA, Crowley WF Jr (2000) Endocrinol Metab 81:32033207
Maffei L, Rochira V, Zirilli L,
Aromatase inhibition in the human Korach KS (1994) Insights from the Antunez P, Aranda C, Fabre B,
male reveals a hypothalamic site of study of animals lacking functional Simone ML, Pignatti E, Simpson
estrogen feedback. J Clin Endocrinol estrogen receptor. Science ER, Houssami S, Clyne CD,
Metab 85:30273035 266:15241527 Carani C (2007) A novel compound
Heard RD, Jellinck PH, ODonnelll VJ Lanfranco F, Zirilli L, Baldi M, heterozygous mutation of the
(1955) Biogenesis of the estrogens: Pignatti E, Corneli G, Ghigo E, aromatase gene in an adult man: a
the conversion of testosterone-4- Aimaretti G, Carani C, Rochira V reinforced evidence on the
C14 to estrone in pregnant mare. (2008) A novel mutation in the relationships among congenital
Endocrinology 57:200204 human aromatase gene: insights on estrogen deficiency, adiposity and

434
Chapter 19: Pathophysiology of estrogen action in men

the metabolic syndrome. Clin not its hypothalamic effects: transdermal estradiol on basal and
Endocrinol 67:218224 evidence from the tandem study of stimulated pituitary hormones. J Clin
normal and gonadotropin-releasing Endocrinol Metab 87:28572862
Mauras N, OBrien KO, Klein KO,
Hayes V (2000) Estrogen hormone-deficient men. J Clin Rochira V, Balestrieri A, Madeo B,
suppression in males: metabolic Endocrinol Metab 93:784791 Spaggiari A, Carani C (2002b)
effects. J Clin Endocrinol Metab Pitteloud N, Dwyer AA, DeCruz S, Congenital estrogen deficiency in
85:23702377 Lee H, Boepple PA, Crowley WF Jr, men: a new syndrome with different
Hayes FJ (2008b) The relative role phenotypes; clinical and therapeutic
Mauvais-Jarvis F (2011) Estrogen and
of gonadal sex steroids and implications in men. Mol Cell
androgen receptors: regulators of
gonadotropin-releasing hormone Endocrinol 193:1928
fuel homeostasis and emerging
pulse frequency in the regulation of
targets for diabetes and obesity. Rochira V, Zirilli L, Madeo B,
follicle-stimulating hormone
Trends Endocrinol Metab 22:2433 Balestrieri A, Granata AR, Carani C
secretion in men. J Clin Endocrinol
(2003) Sex steroids and sexual desire
Mellstrm D, Vandenput L, Metabol 93:26862692
mechanism. J Endocrinol Invest
Mallmin H, Holmberg AH,
Lorentzon M, Odn A,
Raman JD, Schlegel PN (2002) 26:2936
Aromatase inhibitor for
Johansson H, Orwoll ES, Labrie F, male infertility. J Urol Rochira V, Granata ARM, Madeo B,
Karlsson MK, Ljunggren O, Zirilli L, Rossi G, Carani C (2005)
167:624629
Ohlsson C (2008) Older men with Estrogens in males: what have we
low serum estradiol and high serum Raven G, de Jong FH, Kaufman JM, learned in the last 10 years? Asian
SHBG have an increased risk of de Ronde W (2006) In men, J Androl 7:320
fractures. J Bone Miner Res peripheral estradiol levels directly
reflect the action of estrogens at the Rochira V, Zirilli L, Genazzani AD,
23:15521560 Balestrieri A, Aranda C, Fabre B,
hypothalamopituitary level to
Morishima A, Grumbach MM, Simpson inhibit gonadotropin secretion. Antunez P, Diazzi C, Carani C,
ER, Fisher C, Qin K (1995) Aromatase J Clin Endocrinol Metab Maffei L (2006a) Hypothalamic-
deficiency in male and female sibling 91:33243328 pituitary-gonadal axis in two men
caused by a novel mutation and the with aromatase deficiency: evidence
physiological role of estrogens. J Clin Robbins A (1996) Androgens and male that circulating estrogens are
Endocrinol Metab 80:36893699 sexual behavior. Trends Endocrinol required at the hypothalamic level
Metab 7:345350 for the integrity of gonadotropin
Mowa CN, Jesmin S, Miyauchi T
(2006) The penis: a new target and Rochira V, Carani C (2009) Aromatase negative feedback. Eur J Endocrinol
deficiency in men: a clinical 155:513522
source of estrogen in male
reproduction. Histol Histopathol perspective. Nature Reviews Rochira V, Balestrieri A, Madeo B,
21:5367 5:559568 Zirilli L, Granata ARM, Carani C
Rochira V, Faustini-Fustini M, (2006b) Osteoporosis and male
ODonnelll L, Robertson KM, Jones
Balestrieri A, Carani C (2000) age-related hypogonadism: role of
ME, Simpson ER (2001) Estrogen
Estrogen replacement therapy in a sex steroids on bone patho
and spermatogenesis. Endocr Rev
man with congenital aromatase (physiology). Eur J Endocrinol
22:289318
deficiency: effects of different doses 154:175185
Pentakainen V, Erkkila K, of transdermal estradiol on bone Rochira V, Madeo B, Zirilli L,
Suomaleinen L, Parvinen M, Dunkel L mineral density and hormonal Caffagni G, Maffei L, Carani C
(2000) Estradiol acts as a germ cell parameters. J Clin Endocrinol Metab (2007) Oestradiol replacement
survival factor in the human testis. 85:18411845 treatment and glucose homeostasis
J Clin Endocrinol Metab 85:20572067
Rochira V, Balestrieri A, Madeo B, in two men with congenital
Pilgrim C, Reisert J (1992) Differences Baraldi E, Faustini-Fustini M, aromatase deficiency: evidence for a
between male and female brains Granata AR, Carani C (2001) role of oestradiol and sex steroids
developmental mechanisms and Congenital estrogen deficiency: in imbalance on insuline sensitivity in
implications. Horm Metab Res search of the estrogen role in men. Diabet Med 24:14911495
24:353359 human male reproduction. Mol Cell Rochira V, Madeo B, Zirilli L, Carani C
Pitteloud N, Dwyer AA, DeCruz S, Endocrinol 178:107115 (2009) Estrogens and male
Lee H, Boepple PA, Crowley WF Jr, Rochira V, Balestrieri A, Faustini- reproduction. In: de Groot L,
Hayes FJ (2008a) Inhibition of Fustini M, Borgato S, Beck-Peccoz P, McLachlan RI (eds) Endocrinology
luteinizing hormone secretion by Carani C (2002a) Pituitary function of Male Reproduction. www.
testosterone in men requires in a man with congenital aromatase endotext.org/male/male17/
aromatization for its pituitary but deficiency: effect of different doses of maleframe17.htm (MDText.com,

435
Chapter 19: Pathophysiology of estrogen action in men

Inc), South Dartmouth, MA, Simpson ER, Mahendroo MS, Means Vennelli GB, Granchi S, Orlando C,
Chapter 17 GD, Kilgore MW, Hinshelwood Gelmini S, Ledda F, Forti G, Maggi
Roselli CE, Larkin K, Resko JA, MM, Graham-Lorence S, M (2004) Oxytocin receptor is
Stellflug JN, Stormshak F (2004) Amarneh B, Ito Y, Fisher CR, expressed in the penis and mediates
The volume of a sexually Michael MD (1994) Aromatase an estrogen-dependent smooth
dimorphic nucleus in the ovine cytochrome P450, the enzyme muscle contractility. Endocrinology
medial preoptic area/anterior responsible for estrogen 145:18231834
hypothalamus varies with sexual biosynthesis. Endocr Rev 15:342355 Wickman S, Dunkel L (2001)
partner preference. Endocrinology Smith EP, Boyd J, Frank GR, Inhibition of P450 aromatase
145:478483 Takahashi H, Cohen RM, enhances gonadotropin secretion in
Roselli CE, Liu M, Hurn PD (2009) Specker B, Williams TC, Lubahn early and midpubertal boys:
Brain aromatization: classic roles DB, Korach KS (1994) Estrogen evidence for a pituitary site of action
and new perspectives. Semin Reprod resistance caused by a mutation in of endogenous E. J Clin Endocrinol
Med 27:207217 the estrogen-receptor gene in a Metab 86:48874894
man. N Engl J Med 331: Wilcox AJ, Baird DD, Weinberg CR,
Roselli CE, Estill CT, Stadelman HL,
10561061 Hornsby PP, Herbst AL (1995)
Meaker M, Stormshak F (2011)
Separate critical periods exist for Taxel P, Kennedy DG, Fall PM, Willard Fertility in men exposed prenatally
testosterone-induced differentiation AK, Clive JM, Raisz LG (2001) The to diethylstilbestrol. N Engl J Med
of the brain and genitals in effect of aromatase inhibition on sex 332:14111416
sheep. Endocrinology steroids, gonadotropins, and Wright CL, Schwarz JS, Dean SL,
152:24092415 markers of bone turnover in older McCarthy MM (2010) Cellular
Saez JM (1994) Leydig cells: endocrine, men. J Clin Endocrinol Metab mechanisms of estradiol-mediated
paracrine, and autocrine regulation. 86:28692874 sexual differentiation of the brain.
Endocr Rev 15:574626 Toppari J, Skakkebaek NE (1998) Trends Endocrinol Metab
Saylam B, Efesoy O, Cayan S (2011) Sexual differentiation and 21:553561
The effect of aromatase inhibitor environmental endocrine Zachmann M, Prader A, Sabel AH,
letrozole on body mass index, disruptors. Baillires Clin Crigler JF, Ritzen MF, Atares M,
serum hormones, and sperm Endocrinol Metab 12:143155 Ferrandez AF (1986) Pubertal
parameters in infertile men. Fertil Toppari J, Larsen JC, Christiansen P, growth in patients with androgen
Steril 95:809811 Giwercman A, Grandjean P, insensitivity: indirect evidence for
Sharpe RM (1998) The roles of Guillette LJ Jr, Jgou B, Jensen TK, the importance of estrogens in
oestrogen in the male. Trends Jouannet P, Keiding N (1996) Male pubertal growth of girls. J Pediatr
Endocrinol Metab 9:371377 reproductive health and 108:694697
environmental xenoestrogens. Zirilli L, Rochira V, Diazzi C,
Simoni M, Rochira V, Faustini-Fustini Environ Health Persp 104:741803
M, Carani C (1999) Estrogen Caffagni G, Carani C (2008)
resistance and aromatase deficiency. Vandenput L, Ohlsson C (2010) Sex Human models of aromatase
In: Beck-Peccoz P (ed) Regulation of steroid metabolism in the regulation deficiency. J Steroid Biochem Molec
Pituitary Hormone Secretion, of bone health in men. J Steroid Biol 109:212218
HypoCCS Series, Vol. 3. Biochem Mol Biol 121:582588 Zondek B (1934) Mass excretion of
BioScientifica Ltd, Bristol, Vignozzi L, Filippi S, Luconi M, oestrogenic hormone in the urine of
pp 8595 Morelli A, Mancina R, Marini M, the stallion. Nature 33:209210

436
Chapter
Dehydroepiandrosterone and

20 androstenedione
Bruno Allolio, Wiebke Arlt, and Stefanie Hahner

20.1 Introduction 437 20.6 Androstenedione administration in


20.2 Dehydroepiandrosterone secretion and clinical studies 446
age 438 20.7 The emerging therapeutic profile of
20.3 Epidemiology 438 DHEA 447
20.4 Mechanisms of action 439 20.7.1 Effects on the central nervous
20.4.1 Dehydroepiandrosterone 439 system 447
20.4.2 Androstenedione 441 20.7.2 Metabolism and body
20.5 Treatment with DHEA: clinical studies 442 composition 447
20.5.1 Patients with adrenal insufficiency 442 20.7.3 Skeletal system 448
20.5.2 Elderly subjects 444 20.7.4 Skin 449
20.5.3 Patients with impaired mood and 20.7.5 Immune system 449
well-being 445 20.8 Practical approach to the patient with DHEA
20.5.4 Patients with immunological deficiency 449
disorders 445 20.9 Future perspectives 449
20.5.5 Dehydroepiandrosterone 20.10 Key messages 450
supplementation in diminished ovarian 20.11 References 451
reserve 446

20.1 Introduction the degenerative changes observed in human aging


Man, together with higher primates, has adrenals and that administration of DHEA may reverse some of
secreting large amounts of dehydroepiandrosterone these changes. Moreover, the still ongoing availability of
(DHEA) and its sulfate ester, DHEAS. The physio- DHEA as a food supplement in the USA and its
logical role of these steroid hormones is still not fully marketing as an anti-aging drug resulted in large scale
understood. However, in recent years a growing self-administration without medical supervision.
number of careful investigations have helped to better However, in rodents circulating levels of DHEA and
appreciate the function of DHEA(S). DHEAS are several orders of magnitude lower than in
Dehydroepiandrosterone is distinct from the two humans and no age-related decline in DHEA concen-
other major adrenocortical steroids cortisol and trations has been documented. This indicates that experi-
aldosterone in declining with advancing age. More- mental studies in laboratory animals receiving high doses
over, administration of DHEA to experimental animals of DHEA have little bearing for human physiology.
has demonstrated a multitude of beneficial effects on the This chapter, therefore, will focus mainly on
prevention of cancer, heart disease, diabetes and obesity data generated in humans. Dehydroepiandrosterone
(Svec and Porter 1998). This has led to the assumption (sulfate), or DHEA(S), will refer to both DHEA and
that the age-related decline of DHEA may play a role in DHEAS. In addition, clinical studies concerning

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

437
Chapter 20: Dehydroepiandrosterone and androstenedione

DHEAS (nmol/l) of cortisol, while DHEAS does not vary throughout


Birth
the day. There is a clear gender difference in DHEA(S)
10 000 concentrations, with lower DHEAS concentrations in
adult women compared to men (Orentreich et al.
1984). The physiological basis for this gender differ-
5000
ence is not fully clear. Some of the circulating DHEA
in males may be contributed by the testes (Nieschlag
et al. 1973). Roth et al. (2011) demonstrated that both
DHEA and androstenedione are highly concentrated
0 (27-fold and 175-fold, respectively) in the human
10 20 30 40 50 60 70 testes, and respond to hCG. However, substantial
Age (years) changes in intratesticular DHEA or androstenedione
Fig. 20.1 Serum DHEAS concentrations during the human were not reflected by the circulating concentrations of
life cycle. these steroids. No contribution from the ovaries has
been reported, although they may indirectly affect
androstenedione, another less widely used steroid
DHEA(S) levels (Cumming et al. 1982).
hormone precursor, will also be covered.
There is also a clear genetic component to circu-
20.2 Dehydroepiandrosterone lating DHEA(S) levels, which show high interindivid-
ual variability and vary significantly in populations of
secretion and age different ethnic origins (Spencer et al. 2007).
In humans and in some non-human primates, the
secretion of DHEA(S) shows a characteristic pattern
throughout the life cycle (Orentreich et al. 1984; 20.3 Epidemiology
Fig. 20.1). Dehydroepiandrosterone (sulfate) is There is some gender effect of high DHEAS levels in
secreted in high quantities by the fetal zone of the epidemiological studies: an inverse correlation
adrenal cortex, leading to high circulating DHEAS between DHEAS levels and death from any cause
levels at birth. As the fetal zone involutes, a sharp fall was reported for men (>50 years of age) but not for
in serum DHEA(S) concentrations is observed post- women (Barrett-Connor and Goodman-Gruen 1995).
partum to almost undetectable levels after the first In a prospective cohort study in 622 subjects of 65
months of life. Levels remain low until they gradually years and older, mortality at two and four years was
increase between the 6th and 10th years of age; associated with low serum DHEAS at baseline in men
a phenomenon termed adrenarche (Sklar et al. 1980; but not in women (Berr et al. 1996). Similarly in a
Auchus and Rainey 2004). Recently it has been sug- report including men (n = 963) and women (n = 1171)
gested that a rise in intra-adrenal cortisol during >65 years of age, all cause and cardiovascular disease
childhood adrenal growth may lead to inhibition mortality were highest in the lowest DHEAS quartile
of 3b-hydroxysteroid dehydrogenase type 2 and for men. Again no significant association of circulat-
thereby contribute to the initiation of adrenarche ing DHEAS and mortality was found for women
(Topor et al. 2011). Dehydroepiandrosterone (sulfate) (Trivedi and Khaw 2001). In addition, Mazat et al.
concentrations peak during the third decade, followed (2001) found no association between mortality and
by a steady decline with advancing age, so that levels DHEAS levels in women; whereas in men the relative
during the eighth and ninth decade are only 1020% risk of death was 1.9 (p < 0.01) for those with the
of those in young adults (Orentreich et al. 1992). This lowest concentrations of DHEAS. However, in a more
decline has been termed adrenopause in spite of recent study in 270 postmenopausal women undergo-
unchanged or even increased cortisol secretion during ing coronary angiography for suspected CAD, lower
aging (Laughlin and Barrett-Connor 2000). The age- DHEAS concentrations were predictive of higher car-
related decline in DHEA(S) levels shows high diovascular and all-cause mortality (Shufelt et al.
interindividual variability (Spencer et al. 2007), and 2010). As DHEAS was related to severity of CAD, it
is associated with a reduction in size of the zona was suggested that low DHEAS was mechanistically
reticularis (Parker et al. 1997). Dehydroepiandroster- linked to atherosclerosis. Furthermore, in 302 post-
one secretion follows a diurnal rhythm similar to that menopausal women hospitalized for stroke, DHEAS

438
Chapter 20: Dehydroepiandrosterone and androstenedione

Fig. 20.2 Bioconversion of DHEA and


DHEA-ST androstenedione to sex steroids.
Abbreviations: 3b-HSD, 3b-hydroxysteroid
DHEA DHEAS dehydrogenase activity; 17b-HSD,
17b-hydroxysteroid dehydrogenase
STS activity; STS, steroid sulfatase activity;
3b-HSD DHEA-ST, DHEA sulfotransferase activity
(mainly mediated by SULT2A1).

Aromatase
Androstenedione Estrone

17b-HSD 17b-HSDs

Aromatase
Testosterone 17-Estradiol

5a-Reductase

5-Dihydrotestosterone

was inversely related to stroke severity; whereas the androstenedione by the activity of 3b-hydroxysteroid
outcome (handicap, death) was directly associated with dehydrogenase (3b-HSD) and then further converted to
DHEAS and androstenedione levels. The sex difference testosterone and estradiol by isoenzymes of 17b-hydro-
observed in many (but not all) studies may be related, xysteroid dehydrogenase (17b-HSD) and P450 aroma-
in part, to sex-specific differences in bioconversion of tase, respectively (Fig. 20.2). Only lipophilic DHEA can
DHEA(S) (Arlt et al. 1998; 1999a; see below). be converted intracellularly to androgens and estro-
In addition, low DHEAS levels may be a non- gens. Thus the local availability and activity of DHEA
specific marker of poor health status and thereby asso- sulfotransferase (SULT2A1) and steroid sulfatase
ciated with an increased risk of severe illness and death. determines the ratio of DHEA activation (via conver-
Low DHEA(S) concentrations have been found in SLE, sion to sex steroids) to inactivation (via secretion as
dementia, breast cancer and rheumatoid arthritis, and DHEAS back into the circulation). It has consistently
there is an inverse relationship between serum DHEAS been shown that oral DHEA is readily converted to
levels and severity of disease (Deighton et al. 1992). both DHEAS and downstream steroids (e.g. androste-
Thus, low DHEAS levels may indicate the presence of a nedione) (Arlt et al. 1998; 1999a). Accordingly, a
not yet apparent disease, which determines a future genome-wide association study in 14 846 subjects
risk of morbidity or even mortality. found an association of serum DHEAS with poly-
morphisms in the SULT2A1 gene (Zhai et al. 2011).
20.4 Mechanisms of action It has also long been assumed that DHEAS undergoes
continuous conversion to DHEA; thereby serving as a
20.4.1 Dehydroepiandrosterone large circulating pool for DHEA generation (Allolio
Three mechanisms of action have been described for and Arlt 2002). However, this concept has been chal-
DHEA: as a precursor for active sex steroids, as a neuro- lenged by the results of intravenous administration of
steroid interacting with receptors in the central nervous DHEAS in healthy men (Hammer et al. 2005). While
system, and as a ligand for a specific DHEA receptor. DHEAS is generated from circulating DHEA via
As the human steroidogenic enzyme P450c17 hepatic SULT2A1 activity in relevant amounts, only
converts almost no 17a-hydroxyprogesterone to minute amounts of circulating DHEA arise from
androstenedione, the biosynthesis of virtually all sex DHEAS, indicating that hepatic steroid sulfatase
steroids begins with the conversion of 17-hydroxypreg- hardly contributes to circulating DHEA. This observa-
nenolone to DHEA. Thus, only DHEA is converted to tion has profound implications, as it is now clear that

439
Chapter 20: Dehydroepiandrosterone and androstenedione

changes in circulating DHEAS may not always reflect testosterone levels (Arlt et al. 1999a). As ADG is the
changes in DHEA availability. This has been convin- major metabolite of DHT (Giagulli et al. 1989), it
cingly demonstrated in patients with septic shock who reflects increased DHT generation in peripheral
have low circulating DHEAS in the presence of high androgen target tissues. Furthermore, DHEA can
DHEA concentrations (Arlt et al. 2006). Moreover, also be converted to hydroxylated metabolites like
inactivating mutations in the cofactor enzyme PAPS androst-5-ene-3b,7b,17b-triol (bAET), which has
synthase 2, that provides the sulfate donor PAPS to anti-inflammatory and cholesterol-lowering activity
human DHEA sulfotransferase, have been shown to (Stickney et al. 2011). The potential clinical role of
result in non-detectable DHEAS but high levels of this and other steroids derived from the prohormone
DHEA and downstream androgens (Noordam et al. DHEA awaits further studies.
2009). This finding has provided definitive evidence In addition, DHEA is considered a neurosteroid.
for a crucial role of DHEA sulfation in regulating the There is compelling evidence for DHEA synthesis and
production of active androgens. The situation is dif- action in the central nervous system. Several studies
ferent in pregnancy, as here DHEAS can be converted have demonstrated the synthesis of P450c17 and
to DHEA due to abundant placental expression of other key steroidogenic enzymes in the brain (Zwain
steroid sulfatase (Reed et al. 2005). Furthermore, the and Yen 1999), thereby providing the tools to gener-
situation may also be different in various target ate DHEA in the absence of adrenal and gonadal
tissues (e.g. breast, prostate) where substantial steroid function. Dehydroepiandrosterone influences neur-
sulfatase activity may lead to the local generation of onal activity via interaction with various receptors
relevant amounts of DHEA and consecutively down- (N-methyl-D-aspartate (NMDA) receptor, sigma
stream sex steroids not reflected in the circulation receptor, g-aminobutyric acid (GABAA) receptor
(Stanway et al. 2007). (Perez-Neri et al. 2008)). Dehydroepiandrosterone
The widespread presence of 3b-HSD, 17b-HSD, may also influence brain function by direct binding
5a-reductase and P450 aromatase results in almost to dendritic brain microtubule-associated protein
ubiquitous peripheral generation of sex steroids from MAP2C (Laurine et al. 2003). Animal and in-vitro
DHEA. Tissues involved include liver, skin, prostate, studies have shown that DHEA(S) affects neuronal
bone, breast and brain. It has been estimated that 40% growth and development and improves glial survival,
of the androgen pool in elderly men and 50100% learning and memory (Compagnone and Mellon
of estrogen synthesis in pre- and postmenopausal 1998; Svec and Porter 1998). An intriguing observa-
women results from adrenal steroids converted in tion is the direct interaction of DHEA with nerve
peripheral tissues (Labrie 2010). growth factor (NGF) receptors. This binding of
The concept of peripheral synthesis, action and DHEA to NGF receptors is functional, mediating
metabolism of steroid hormones from inactive pre- anti-apoptotic effects and providing a mechanism of
cursors within the same target cell has been coined its neuroprotective action (Lazaridis et al. 2011).
intracrinology. Such intracrine processes are diffi- Third, there is growing evidence for DHEA action
cult to study, as serum parameters may only partially outside the brain via specific receptors, although such
reflect target cell physiology. Nonetheless there a specific DHEA receptor has not yet been fully char-
is evidence that the bioconversion of DHEA(S) acterized or even cloned. High-affinity binding sites
follows a sexually dimorphic pattern, with preferential for DHEA have been described in murine and human
increases in androgenic activity in women and T-cells (Meikle et al. 1992; Okabe et al. 1995), but
increases in circulating estrogens in men (Arlt et al. their specificity for DHEA remained questionable.
1998; 1999a). However, in men with combined High-affinity binding sites for DHEA were identified
adrenal insufficiency and hypogonadism due to in bovine endothelial cells (Liu and Dillon 2002).
hypopituitarism, oral DHEA administration induces In these cells DHEA activates eNOS via a G-protein-
significant increases in both estrogens and androgens coupled plasma membrane receptor. Similarly,
(Young et al. 1997). Moreover, after oral administra- DHEA affects extracellular-signal-regulated kinase 1
tion of DHEA to men a pronounced increase in (ERK-1) phosphorylation in human vascular smooth
circulating 5a-androstane-3a,17b-diol-glucuronide muscle cells, independently of androgen and estrogen
(ADG) is found, indicating increased peripheral receptors (Williams et al. 2002). Dehydroepiandros-
androgen synthesis not reflected by changes in circulating terone also causes vascular endothelial proliferation

440
Chapter 20: Dehydroepiandrosterone and androstenedione

200
180 30
Androstenedione (nmol/l)

160
25

Testosterone (nmol/l)
140
120 20
100
15
80
60 10
40
5
20
0 0

0 2 4 6 8 22 24 26 0 2 4 6 8 22 24 26
Time (h) Time (h)
Fig. 20.3 Increase of testosterone in healthy young women after oral administration of 100 mg androstenedione (Kicman et al. 2003).

in a MAP-kinase dependent manner (Liu et al. 2008). Taken together, there is clear evidence that DHEA
It was shown in human and bovine vascular endothe- has a complex and specific activity profile, which in
lial cells that DHEA leads to a significant production part is gender specific due to its sex-related differen-
of H2O2 at physiological concentrations via specific tial pattern of downstream bioconversion to potent
membrane receptors. Furthermore, DHEA-induced sex steroids. While an interaction with several known
endothelial cell proliferation was blocked by catalase, receptors has been convincingly shown, a specific
suggesting that H2O2 is a key molecule mediating the DHEA receptor remains to be characterized.
proliferative response to DHEA (Iruthayanathan et al.
2011). In vascular smooth muscle cells, DHEA affects
phosphorylation and translocation of the transcrip- 20.4.2 Androstenedione
tion factor FoxO1, possibly by indirectly modifying Androstenedione is not only a product of DHEA
mineralocorticoid receptor activity (Lindschau et al. metabolism, but may be regarded as a prohormone
2011). Interaction with sigma receptors may also itself. It can be converted to testosterone by 17b-
play a role outside the brain, as DHEA was shown HSDs or to estrone by the aromatase enzyme complex
to upregulate and stimulate sigma-1 receptors in the (Leder et al. 2000). Accordingly, administration
aorta and the kidney in experimental animals, indu- of androstenedione may alter circulating steroid
cing eNOS activity (Bhuiyan and Fukunaga 2010; hormone concentrations. In women, pronounced
Bhuiyan et al. 2011). These observations strengthen increases not only in circulating androstenedione
the concept of direct and specific hormonal activity of but also in testosterone, DHT and free androgen
DHEA independent of its bioconversion to other index have been described following administration
steroids. Of note, a recent study has provided evi- of 100 mg androstenedione (Kicman et al. 2003; Bas-
dence for a distinct action of DHEAS in human sindale et al. 2004; Fig. 20.3). In contrast, in men the
neutrophils: DHEAS, but not unconjugated DHEA, effects of oral androstenedione have been variable: in
was found to increase human neutrophil superoxide some trials serum total testosterone concentrations
generation, a crucial mechanism underpinning neu- were not affected by 100 mg androstenedione (King
trophil function, by direct interaction of DHEAS with et al. 1999; Brown et al. 2000). However, 300 mg
protein kinase C (PKC) b (Radford et al. 2010). Of androstenedione induced increases in testosterone
note, human neutrophils were shown to be capable levels (Leder et al. 2000). Importantly, clear increases
of active and specific transmembrane import of in estrogens were observed after oral ingestion of
hydrophilic DHEAS via the organic anion transporter androstenedione in young and elderly men (King
polypeptide D (OATP-D) that appears as selectively et al. 1999; Brown et al. 2000; Leder et al. 2000) and
expressed in neutrophils but no other human blood less pronounced also in women (Brown et al. 2004),
cell (Radford et al. 2010). an effect quite similar to oral DHEA administration.

441
Chapter 20: Dehydroepiandrosterone and androstenedione

However, in postmenopausal women only an increase Hahner et al. 2007; Erichsen et al. 2009). Jakobi
in testosterone and estrone, but not in estradiol was et al. have provided some more insight into the mech-
reported (Leder et al. 2002). The intracrine activation anism of increased fatigue in conventionally treated
of androstenedione similar to DHEA was high- patients with adrenal insufficiency. Muscle function
lighted by a detailed analysis of the metabolism of (twitch tension, central activation) was reduced and
orally administered androstenedione in young men patients self-terminated a submaximal fatigue proto-
(Leder et al. 2001), observing increases in the excre- col significantly earlier than controls (5  1 vs. 10  1
tion rates of conjugated testosterone, androsterone, min, p = 0.006; Jakobi et al. 2001). Recent data fur-
etiocholanolone and dihydrotestosterone. It was con- thermore demonstrate increased mortality in adrenal
cluded that orally administered androstenedione is insufficiency patients, specifically from respiratory
largely metabolized to androgen metabolites before infections (Tomlinson et al. 2001; Bergthorsdottir
release into the general circulation. Thus again the et al. 2006; Bensing et al. 2008; Erichsen et al. 2009).
biological activity of androstenedione is incompletely For replacement of DHEA(S) in adrenal insuffi-
reflected by circulating active sex steroids. Similar ciency, an oral dose of 2550 mg DHEA per day
hormone profiles were obtained using sublingual has consistently been found to restore circulating
androstenediol in young men (Brown et al. 2002). DHEA(S) into the normal range of young adults (Arlt
Clear increases in serum testosterone were found by et al. 1999b; Hunt et al. 2000). Due to the downstream
bypassing first-pass hepatic metabolism using this bioconversion, lasting increases in circulating andro-
sublingual administration. Intriguingly, longer-term gens have been demonstrated in women (Arlt et al.
administration of 200 mg/day androstenedione in 1998; 1999b; Fig. 20.4).
elderly men led to an unexpected increase in circu- So far, several trials have investigated the effect of
lating DHEAS (upstream of androstenedione) and DHEA replacement on well-being and on metabolic
a reduced increase to an acute challenge with andros- parameters in both primary and secondary adrenal
tenedione, suggesting enhanced androstenedione insufficiency (Arlt et al. 1999b; Hunt et al. 2000;
metabolism (Beckham and Earnest 2003). Callies et al. 2001; Johannsson et al. 2002; Lovas
At present there is no evidence that androstene- et al. 2003; Libe et al. 2004; Bilger et al. 2005; van
dione has biological activity independent of its down- Thiel et al. 2005; Brooke et al. 2006; Dhatariya et al.
stream conversion to sex steroids. 2008; Gurnell et al. 2008). Dehydroepiandrosterone
has shown beneficial effects on subjective health
status, mood and sexuality. These effects could also
20.5 Treatment with DHEA: be demonstrated in men who do not suffer from
clinical studies androgen deficiency owing to the preserved androgen
production by their testes, suggesting that these
20.5.1 Patients with adrenal insufficiency effects are in part mediated by neurosteroidal activity
The classical approach to study the physiological role (see also Chapter 17).
of a hormone in humans is to analyze the effect on a In the first double-blind study (Arlt et al. 1999b),
hormonal deficit and the changes induced by replace- treatment with DHEA in women with adrenal insuffi-
ment of the missing hormone. Thus, adrenal insuffi- ciency raised the initially low concentrations of
ciency is the most useful model disease to understand DHEA(S), androstenedione and testosterone into
the clinical activity of DHEA. In adrenal insufficiency, the normal range. Serum concentrations of SHBG,
not only cortisol and (in primary adrenal insuffi- total cholesterol and HDL-C decreased significantly.
ciency) aldosterone is lacking, but also DHEA, and Dehydroepiandrosterone improved well-being and
one might speculate that replacement of cortisol sexuality: compared to placebo DHEA resulted in a
and aldosterone alone is not sufficient to fully restore significant decrease in the scores for depression and
well-being in adrenal insufficiency. Intriguingly, anxiety, as well as for a global severity index of the
it has recently been clearly demonstrated that, despite SCL-90-R questionnaire. Beneficial effects of DHEA
established replacement of glucocorticoids and treatment on anxiety and depression were also
mineralocorticoids, patients suffer from impaired observed for the Hospital Anxiety and Depression
well-being with increased fatigue, depression, loss of Scale (HADS). A reduction in fatigue was evident
libido and impaired vitality (Jakobi et al. 2001; from the Giessen Complaint List. Treatment with

442
Chapter 20: Dehydroepiandrosterone and androstenedione

Testosterone (nmol/l) Fig. 20.4 Lasting increases of serum


testosterone in healthy dexamethasone
(Dex)-suppressed women after oral
3 administration of 100 mg DHEA versus
Dex + 100 mg DHEA placebo (Arlt et al. 1998).
2.5
Dex + placebo

1.5

0.5

0
0 5 10 15 20 25
Time (h)

DHEA resulted in significant increases in the initially patients with primary adrenal insufficiency (n = 106),
low scores of all four visual-analog scales for sexuality. who received 12 months of DHEA replacement (50
Dehydroepiandrosterone did not affect fasting serum mg/day) or placebo in a parallel-group study design
glucose, insulin and parameters of body composition (Gurnell et al. 2008). Results of this phase III trial
(Callies et al. 2001). Using an incremental cycling demonstrated significant improvement of SF-36 role
test, maximum workload was 95.8  20.4 W after emotional function scores and some slight improve-
DHEA, compared to 91.7  24.1 W after placebo ment also in further QoL scores during DHEA
(p = 0.057). replacement. No significant benefit of DHEA treat-
Androgenic skin effects of DHEA treatment were ment on fatigue or cognitive or sexual function could
reported in 19 out of the 24 women but were mostly be demonstrated. In addition they found a reversal of
mild and transient (Arlt et al. 1999b). loss of BMD at the femoral neck by DHEA replace-
Improvement in mood and fatigue was also ment as assessed by DXA. Body composition analysis
observed after DHEA replacement in Addisons by DXA also revealed a significant increase in lean
disease in the trial reported by Hunt et al. (2000). body mass, while fat mass remained unchanged
The hormonal changes induced by DHEA in females (Gurnell et al. 2008).
were virtually identical to those reported by Arlt et al. In 38 women with secondary adrenal insufficiency
(1999b), with increases in serum DHEA(S), androste- due to hypopituitarism, DHEA (2030 mg/day) was
nedione and testosterone into normal range for used (Johannsson et al. 2002). Dehydroepiandrosterone
women. In males, serum testosterone and SHBG or placebo was given for six months in a randomized,
did not change. Hunt et al. (2000) found a significant placebo-controlled, double-blind study, followed by a
increase in self-esteem after DHEA substitution. six-month open treatment period. The percentage of
Using a Profile of Mood State Questionnaire it was partners of the patients who reported improved alert-
demonstrated that evening mood and evening fatigue ness, stamina and initiative in their spouses were 70%,
was improved by DHEA. No changes in BMD, BMI, 64% and 55%, respectively, in the DHEA group, and
serum cholesterol or insulin sensitivity were observed 11%, 6% and 11%, respectively, in the placebo group
after DHEA treatment. As the beneficial effects in (p < 0.05). Sexual relations tended to improve
this study were also observed in male patients who (p = 0.06). An increase in or the reappearance of axillary
exhibited no change in testosterone, it was concluded and/or pubic hair was seen in all women given 30 mg
that DHEA acts directly at the central nervous system DHEA and in 69% of women receiving 20 mg DHEA,
rather than via peripheral conversion to androgens but was not found in women receiving placebo. Glucose
(Hunt et al. 2000). The same group followed up on metabolism and lipoproteins remained unaffected
these results and performed the largest study so far in by DHEA, with the exception of transient decrease

443
Chapter 20: Dehydroepiandrosterone and androstenedione

in HDL-C. These values had significantly increased domains of the General Health Questionnaire after
at 12 months (p < 0.05). Bone markers and BMD six months of DHEA.
remained unchanged. Androgenic skin effects were Besides the results of these randomized trials, evi-
more often seen during DHEA treatment. dence from case reports (Kim and Brody 2001; Wit
The role of DHEA replacement in adolescent and et al. 2001) in adrenal insufficiency is available. Kim
young women was investigated by Bilger et al. (2005). and Brody (2001) have described a 24-year-old female
Five young females with hypopituitarism, aged between with Addisons disease and the complaint of neither
15 and 23 years, received DHEA 50 mg/day in a 12- axillary nor pubic hair growth. Dehydroepiandroster-
month, double-blind, placebo-controlled crossover one was added to the conventional replacement ther-
trial. A better life satisfaction was shown under DHEA apy. Pubic hair growth changed from Tanner stage I
replacement in the Life Situation Survey compared to to Tanner stage III within two years of receiving
placebo (110 vs. 102, p = 0.05). In addition, an improve- DHEA at a final dose of 25 mg daily. Similarly, Wit
ment of maximal oxygen uptake (VO2), and decreased et al. (2001) used oral DHEAS (15 mg/m2) for atrichia
percentage body fat was observed which did not reach pubis in four female adolescents with panhypopitui-
significance in the small group investigated. Similarly, tarism (n = 2) or 17-hydroxylase deficiency (n = 2).
Binder and colleagues demonstrated, in a cohort of 23 They found DHEAS an efficacious treatment, leading
young girls and women (1325 years), that 12 months from atrichia pubis to Tanner stage 45 pubic hair.
of DHEA treatment vs. placebo resulted in significant However, other studies could not find any benefi-
improvement of pubic hair Tanner stage and improved cial effect of DHEA replacement (Lovas et al. 2003;
mood and well-being, specifically of depression and Libe et al. 2004), and a more recent meta-analysis of
anxiety and the global severity index as assessed by the RCTs of the effect of DHEA in adrenal insufficiency
SCL-90-R (Binder et al. 2009). revealed only moderate beneficial effects on health-
Two studies have investigated the additive effect of related QoL and depression (Alkatib et al. 2009).
DHEA to GH replacement in secondary adrenal Effects on anxiety and sexual well-being were also
insufficiency. Van Thiel et al. studied the effects small and did not reach statistical significance, which
of DHEA (50 mg/d) for 16 weeks versus placebo in led to the conclusion that larger clinical trials are
GH- and ACTH-deficient men (n = 15), and postme- needed before the routine use of DHEA in adrenal
nopausal women (n = 16), in a double-blind, placebo- insufficiency can be recommended.
controlled crossover study (van Thiel et al. 2005).
A significant improvement by DHEA in SF-36 vitality
scores in both men and women and in the HADS 20.5.2 Elderly subjects
depression score in women was detected. However, The age-related decline in circulating DHEA(S) has
no differences in fatigue scores, QoL assessment in GH led to a number of randomized trials to assess the
deficiency in adults (QOL-AGHDA) or in the sexual effect of oral DHEA in otherwise healthy elderly
functioning questionnaires could be demonstrated. subjects. In a first double-blind, placebo-controlled
Interestingly, IGF-1 levels increased in women during trial using a crossover design, 13 men and 17 women
DHEA replacement by about 18%. No change in IGF-1 aged 4070 years received either 50 mg DHEA or
levels was observed in men, suggesting that the effect placebo for three months (and vice versa) (Morales
may be related to androgenic effects following normal- et al. 1994). The subjects reported an improvement in
ization of androgen levels in women. This observation well-being using a non-validated questionnaire for
was also made by Brooke et al., resulting in a decrease self-assessment of well-being. No change in insulin
of GH replacement doses (Brooke et al. 2006). In sensitivity and body composition was found (see
this double-blind, placebo-controlled trial, 30 females also Sections 20.7.2 and 20.7.3). Bioavailable IGF-1
and 21 males with hypopituitarism received DHEA increased slightly during DHEA treatment, whereas
50 mg over six months, followed by an open phase HDL-C decreased in women. Short-term (two weeks)
of six months of DHEA treatment. Females showed randomized, double-blind studies by Wolf et al.
improvement in QOL-AGHDA score, in SF-36 (Short (1997; 1998) failed to demonstrate any benefit of
Form 36) social functioning and general health per- DHEA on well-being, mood and cognition. Similarly,
ception after six months of DHEA; whereas men in a double-blind, placebo-controlled crossover
showed improvement in self esteem and depression trial, Arlt et al. (2001) found no effect of DHEA

444
Chapter 20: Dehydroepiandrosterone and androstenedione

(50 mg/day) on mood, well-being and sexuality in 20 be related to a selection bias. In almost all studies,
men aged 5069 years after four months of therapy. only healthy subjects with excellent performance
In another placebo-controlled, randomized crossover status at baseline were included, thereby leaving
trial by van Niekerk et al., no effect of 50 mg/day limited space for further improvement. However,
DHEA for 13 weeks on well-being and cognition was from these studies it can be concluded that age-related
found using a wide range of validated self-assessment low DHEA concentrations do not necessarily lead
questionnaires and standardized test batteries, res- to impaired well-being, cognition and sexuality per
pectively (van Niekerk et al. 2001). No effect of DHEA se (Allolio and Arlt 2002).
on activities of daily living was found after three
months of 100 mg/d DHEA in 39 men aged 6084
years in another placebo-controlled crossover trial
20.5.3 Patients with impaired mood
(Flynn et al. 1999). and well-being
In the DHEAge study, Baulieu et al. (2000) inves- Consistent with the effects of DHEA on mood and
tigated the effects of 50 mg/d DHEA vs. placebo in a well-being in patients with adrenal insufficiency,
double-blind, randomized, parallel study including beneficial effects were also observed in randomized,
140 men and 140 women aged 6079 years. In general double-blind studies in patients with major depres-
the results were disappointing. Neither well-being nor sion (Wolkowitz et al. 1999) and midlife dysthymia
cognition was improved by DHEA using a wide range (Bloch et al. 1999). Dehydroepiandrosterone also
of validated tools. In women > 70 years, gains in improved scores on an activities of daily living scale
BMD were detected and libido was increased, but no in patients with myotonic dystrophy (Sugino et al.
significant changes were observed in men. Further- 1998). Reiter et al. have reported an improvement in
more, no influence on well-being or cognition could erectile function, sexual satisfaction and orgasmic
be demonstrated after 12 months of DHEA treatment. function in 4060-year-old men suffering from ED
So far the longest trial by Nair et al. investigated and receiving 50 mg/d DHEA for six months in a
the effect of DHEA and testosterone on QoL, body randomized, double-blind fashion (Reiter et al. 1999).
composition, physical performance and metabolism To compare the efficacy of DHEA vs. placebo in
in elderly subjects (Nair et al. 2006). The authors Alzheimers disease, 58 patients were randomized to
found no significant effects of DHEA in these healthy six months of treatment with DHEA (100 mg/d) or
elderly individuals with low baseline DHEA levels placebo. A transient effect on cognitive performance
on QoL, body composition, physical performance or narrowly missed significance (Wolkowitz et al. 2003),
insulin sensitivity. possibly because of the small patient sample. Strous
Several studies have investigated the role of DHEA et al. have studied the efficacy of DHEA (100 mg/d)
for treatment of symptoms related to postmenopausal in schizophrenic patients with prominent negative
hormone deficiency (Davis et al. 2011). Dehydro- symptoms (Strous et al. 2003). In a double-blind trial,
epiandrosterone treatment has been shown to enhance a significant improvement in negative symptoms (p
parameters of sexual function in postmenopausal < 0.001), as well as in depressive (p < 0.05) and anx-
women after single-dose treatment with 300 mg iety (p < 0.001) symptoms was seen in individuals
(Hackbert and Heiman 2002) or longer term adminis- receiving DHEA.
tration of an oral dose or vaginal cream (Schmidt It seems noteworthy that the pattern of improve-
et al. 2005; Labrie et al. 2009). Most of these studies, ment observed in these trials closely resembled the
however, administered supraphysiological DHEA changes observed in patients with adrenal insufficiency.
doses. Recent longer term studies with larger sample
sizes using 50 mg/d DHEA showed no improvement 20.5.4 Patients with immunological
in sexual function by DHEA and no change in general
well-being (Nair et al. 2006; Kritz-Silverstein et al. disorders
2008; Panjari et al. 2009). In a number of studies, DHEA supplementation
Taking all studies on DHEA supplementation in has been used to modify immune functions and alter
elderly subjects together, the results show only very the course of immunopathies. Most studies have
limited effects of DHEA compared to placebo. An been performed in patients with SLE, a chronic auto-
important explanation for this lack of efficacy may immune inflammatory disease of unknown etiology

445
Chapter 20: Dehydroepiandrosterone and androstenedione

(van Vollenhoven et al. 1994; Chang et al. 2002). The advantage of the IGF-1 increase induced by DHEA.
concept of using DHEA in the treatment of SLE was Furthermore, animal data have shown that androgen
based on the observation that women are more often action in granulosa cells plays a crucial role for
affected and that androgens and DHEA concentra- follicular development and function by promoting
tions are low in patients with SLE (Lahita et al. preantral growth (Sen and Hammes 2010). In women
1987). Moreover, androgen treatment can modify with DOR, DHEA administration was associated
the disease progression in an animal model of SLE with increasing oocyte and embryo counts (Gleicher
(Melez et al. 1980). After preliminary evidence of a et al. 2010). Cohort studies in 89 DOR patients sup-
glucocorticoid-sparing effect of DHEA in patients plemented with DHEA and 101 controls indicated
with mild SLE (van Vollenhoven et al. 1994), a ran- shorter time to pregnancy and higher pregnancy
domized, double-blind, placebo-controlled trial was rates (28.1 vs. 10.9%; Barad et al. 2007). The same
performed (van Vollenhoven et al. 1999). It demon- group reported that DHEA supplementation reduced
strated beneficial effects of DHEA on patient and embryo aneuploidy in a case control study of 22
physician overall assessment, SLE disease activity consecutive patients (Gleicher et al. 2010). In a first
index (SLEDAI) and glucocorticoid requirements. randomized, open trial in 33 women with DOR,
This was confirmed in recent double-blind, random- DHEA (75 mg/d) was associated with a significantly
ized, placebo-controlled trials demonstrating that higher birth rate (23.1 vs. 4%, p < 0.05; Wiser et al.
DHEA (200 mg/d) was well tolerated, reduced the 2010). However, this sample size is rather small and
number of SLE flares, reduced disease activity and the statistical evaluation of this trial has been heavily
allowed reduction of the dosage of glucocorticoids criticized (Kolibianakis et al. 2011). Despite the still
(Chang et al. 2002; Hartkamp et al. 2010). It is weak overall evidence due to the current lack of rig-
important to note that most of these studies included orous randomized trials of sufficient size (Sunkara
women only, and that it remains unclear whether and Coomarasamy 2011; Yakin and Urman 2011), it
similar results can be obtained in men. In a phase II is estimated that about one-third of all IVF centers
uncontrolled pilot trial, DHEA (200 mg/d) was today use DHEA supplementation in women with
effective and safe in patients with refractory Crohns DOR (Gleicher and Barad 2011). Reported side-
disease and ulcerative colitis (Andus et al. 2003). effects were generally mild and included oily skin,
However, to date no placebo-controlled trials have acne and hair loss.
been performed in inflammatory bowel disease. In
all these trials, side-effects were mild, with acne 20.6 Androstenedione
being the most frequently seen adverse event despite
the use of undoubtedly supraphysiological DHEA administration in clinical studies
doses (200 mg/d). Effects of oral androstenedione have not been studied
DHEA supplementation has also been used to in women and have been largely disappointing
enhance the antibody response to tetanus and influ- in men. Short-term (five days) androstenedione
enza vaccines (Danenberg et al. 1997). However, in (100 mg/d) administration had no anabolic effect on
these randomized, placebo-controlled trials, no con- muscle protein metabolism in eugonadal young men
sistent effect of DHEA on protective antibody titers (Rasmussen et al. 2000). In 3056-year-old men,
was found. androstenedione (3  100 mg/d) for 28 days slightly
reduced HDL-C without affecting PSA, suggesting
some androgenic activity (Brown et al. 2000). Serum
20.5.5 Dehydroepiandrosterone HDL-C was also reduced in an eight-week random-
supplementation in diminished ized trial in 20 young men receiving oral androstene-
dione (300 mg/d) (King et al. 1999). Androstenedione
ovarian reserve failed to enhance muscle adaptation to resistance
Dehydroepiandrosterone supplementation for dimin- training in this population (King et al. 1999).
ished ovarian reserve (DOR) in patients undergoing At present, both treatment duration and sample
artificial reproductive treatment has first been sug- sizes have been too limited to draw any firm conclu-
gested by (Casson et al. 2000) to compensate for sions on the clinical efficacy and the ergogenic activ-
adrenocortical changes in aging women and to take ity of androstenedione. However, the profound

446
Chapter 20: Dehydroepiandrosterone and androstenedione

increases in circulating testosterone observed in Libido and sexual satisfaction are influenced by
women after oral androstenedione deserve attention DHEA in women with adrenal insufficiency (Arlt
and should preclude its use as a food supplement et al. 1999b) and in elderly women with age-related
(Kicman et al. 2003; Bassindale et al. 2004). Even low DHEAS (Baulieu et al. 2000). Also in men, only
more so as androstenedione currently belongs to the impaired sexuality benefits from DHEA adminis-
popular supplements in young athletes (Calfee and tration (Reiter et al. 1999), while normal baseline
Fadale 2006) and its abuse is not easily detectable performance cannot be enhanced (Arlt et al. 2001).
(Brown et al. 2004). The effect of DHEA on libido and sexuality is most
likely a consequence of increased androgenic activity
20.7 The emerging therapeutic derived from DHEA by peripheral bioconversion.
In recent years it has become increasingly clear that
profile of DHEA androgens play a key role for female sexuality
20.7.1 Effects on the central (Shifren et al. 2000; Arlt 2003). In fact, the adrenals
are a major source of female androgens (Labrie et al.
nervous system 2003), and their fundamental role for female sexua-
Improvement in mood and well-being have con- lity (Waxenberg et al. 1959) has been rediscovered
sistently been observed in patients with adrenal by studies on the therapeutic potential of DHEA.
insufficiency (Arlt et al. 1999b; Hunt et al. 2000; The available evidence and the superior pharmacoki-
Johannsson et al. 2002) and in patients with depres- netic properties make DHEA a highly attractive tool
sive disorders (Bloch et al. 1999; Wolkowitz et al. for treatment of impaired sexuality in women. How-
1999) and schizophrenia (Strous et al. 2003); ever, firm conclusions must await the results of fur-
particularly improving symptoms of anxiety and ther trials.
depression and their physical correlates. It is import-
ant to note that improvements have only been
observed in subjects with impaired mood and well- 20.7.2 Metabolism and body composition
being at baseline, and that DHEA-induced improve- The effects of DHEA on metabolic parameters (e.g.
ments led to scores in the range of normal healthy lipids, insulin sensitivity) and body composition are
subjects. This indicates that DHEA may normalize mostly not consistent and largely unimpressive.
impaired well-being but will not lead to supranor- However, consistently observed in response to
mal well-being in otherwise healthy subjects (irre- DHEA treatment in both hypoadrenal patients and
spective of the presence of low endogenous DHEAS elderly subjects is an increase in IGF-1 levels (Morales
concentrations). et al. 1998; Villareal et al. 2000; van Thiel et al. 2005;
Several cases of mania have been reported with Brooke et al. 2006; Jankowski et al. 2006; von Muhlen
DHEA treatment (Kline and Jaggers 1999; Markowitz et al. 2008; Weiss et al. 2009). Effects on lipids also
et al. 1999), and we also have observed a similar case appear to be relatively robust. The first study in patients
in a woman with adrenal insufficiency receiving a with adrenal insufficiency already demonstrated a
daily dose of 25 mg DHEA; although a direct causal significant decrease of total cholesterol and HDL-C.
role for DHEA is difficult to establish. Similarly, in the study by Dhatariya et al. (2005) or
The basis for the anxiolytic and antidrepressive Srinivasan et al. (2010), DHEA additionally reduced
activity of DHEA remains to be elucidated but may total cholesterol, LDL-C and triglycerides and also
be related to both androgenic effects and neuroster- HDL-C in hypoadrenal patients. The overall DHEA
oidal actions of DHEA. effect on the lipid profile is modest, mainly consisting
In contrast, there is little evidence that DHEA of a HDL-C-lowering effect (Barnhart et al. 1999;
affects memory or cognition. Negative results have Dayal et al. 2005; Nair et al. 2006; Srinivasan et al.
been found not only in healthy elderly subjects 2010).The mechanisms for the reduction in HDL
(Baulieu et al. 2000) but also in adrenal insufficiency and total cholesterol are most likely mediated by the
(Arlt et al. 2000). Moreover, in Addisons disease effects of androgens on increasing hepatic lipase
cognition is not impaired despite severe endogenous activity, thus impairing hepatic cholesterol formation.
DHEA deficiency (Arlt et al. 2000). Thus it is unlikely The majority of studies assessing the effect of oral
that cognition is a major target of DHEA action. DHEA on insulin sensitivity document no effect

447
Chapter 20: Dehydroepiandrosterone and androstenedione

on insulin sensitivity in healthy elderlies receiving another study in 31 postmenopausal women receiv-
replacement doses of DHEA (Morales et al. 1994; ing 12 weeks of DHEA 50 mg/d in addition to
Casson et al. 1995; Yen et al. 1995; Morales et al. endurance and resistance exercise training; whereas
1998; Nair et al. 2006; Basu et al. 2007; Igwebuike exercising per se significantly increased insulin sen-
et al. 2008; Panjari et al. 2009). Villareal et al. investi- sitivity and improved body composition (Igwebuike
gated 28 women and 28 men aged 6578 years with et al. 2008). Thus, at present a significant effect of
age-related decrease in DHEA levels. The insulin area DHEA on muscular function and body composition
under the curve during the oral glucose tolerance test remains uncertain.
was significantly reduced after six months of DHEA
therapy compared with placebo, and a significant
increase in the insulin sensitivity index in response 20.7.3 Skeletal system
to DHEA was observed (Villareal and Holloszy 2004). Dehydroepiandrosterone replacement has been
In a study in 28 hypoadrenal women receiving 50 reported to increase BMD in elderly subjects. How-
mg oral DHEA over 12 weeks, Dhatariya et al. also ever, small sample size and short duration of treat-
observed lower fasting insulin and glucagon levels ment precluded clear conclusions in many trials.
with DHEA. Insulin sensitivity, assessed by using a Jankowski et al. investigated 70 women and 70 men
hyperinsulinemic-euglycemic clamp, was increased aged 6088 years (Jankowski et al. 2006). They
during DHEA administration (Dhatariya et al. observed an increase in hip BMD in both sexes
2005). However, no effects of DHEA on fasting glu- and, in women, also an increase in lumbar spine
cose, insulin, or glucoseinsulin ratio were observed BMD after one years treatment with 50 mg/d DHEA
in other studies in patients with adrenal insufficiency (Jankowski et al. 2006). Changes in BMD were asso-
(Callies et al. 2001; Libe et al. 2004). ciated with estradiol levels and free estrogen index at
Body composition remains mostly unaffected by 12 months, suggesting that the effects were primarily
DHEA. Morales et al. reported an increase in muscle mediated by increases in serum estradiol (Jankowski
strength in men but not in women, aged 5060 years, et al. 2008). Nair et al. performed a two-year, placebo-
after six months of treatment with 100 mg/d DHEA controlled, randomized, double-blind study involving
(Morales et al. 1998). Application of a 10% DHEA 87 elderly men and 57 elderly women with low levels
cream to the skin daily over 12 months resulted in of sulfated DHEA (Nair et al. 2006). Men receiving
an increase in femoral muscle area in 15 women aged DHEA 75 mg/d had an increase in BMD at the
6070 years (Diamond et al. 1996). A decrease in fat femoral neck. Women who received DHEA 50 mg/d
mass and an increase in fat-free mass assessed by DXA had an increase in BMD at the ultradistal radius.
measurement were furthermore observed by Villareal Several other RCTs reported similar observations,
et al. (2000). However, no effect on body composition with slight increases in BMD and changes in markers
could be observed in several other large trials (Arlt of bone turnover, mainly in women (Labrie et al.
et al. 2001; Percheron et al. 2003; Jankowski et al. 1997; Baulieu et al. 2000; Villareal et al. 2000; von
2006; Nair et al. 2006; von Muhlen et al. 2008). Muhlen et al. 2008).
The possibility of potentiating the response to In patients with adrenal insufficiency, DHEA
exercise training has been investigated. Villareal reversed ongoing loss of BMD at the femoral neck
et al. analyzed the effects of 10 months of DHEA but not at other sites in women receiving DHEA
50 mg/d in combination with weightlifting exercise replacement (Gurnell et al. 2008). However, other
training during the last four months of the study studies could not detect relevant effects on bone mass
(DHEA exercise group, n = 29; placebo exercise (Hunt et al. 2000; Johannsson et al. 2002).
group, n = 27) in 28 women and 28 men aged 6578 Dehydroepiandrosterone effects on bone
years. Dehydroepiandrosterone alone for six months markers were missing in men (Baulieu et al. 2000;
did not significantly increase strength or thigh Arlt et al. 2001; Kahn et al. 2002) and were variable
muscle volume. However, DHEA supplementation in women with either increases, decreases or no
potentiated the effect of four months of weightlifting change in bone resorption markers (Baulieu et al.
training on muscle strength, and on thigh muscle 2000; Villareal et al. 2000; Callies et al. 2001) and
volume, measured by MRI (Villareal and Holloszy increases or no change in osteocalcin (Baulieu et al.
2006). This potentiation was, however, not seen in 2000; Callies et al. 2001).

448
Chapter 20: Dehydroepiandrosterone and androstenedione

At present it seems likely that beneficial effects of 20.8 Practical approach to the patient
DHEA on BMD are small compared to other treat-
ment options and restricted to women, possibly due with DHEA deficiency
to androgenic and estrogenic biotransformation of At present there is still no established indication
DHEA. Again, only large, prospective, controlled and no generally accepted pharmacological prepar-
trials will settle this issue. ation of DHEA for treatment. However, there is
growing evidence (Achermann and Silverman 2001;
Allolio et al. 2007; Alkatib et al. 2009) that DHEA
20.7.4 Skin replacement in patients with adrenal insufficiency is
Skin is an important target of DHEA action: beneficial in a significant percentage of cases. In these
DHEA increases sebum secretion and skin hydra- patients impaired well-being including depression,
tion (Labrie et al. 1997; Baulieu et al. 2000), and reduced vitality and increased fatigue (Hahner et al.
has been reported to reduce facial skin pigmen- 2007) has been documented: symptoms that are likely
tation (yellowness) in elderlies (Baulieu et al. to respond to DHEA replacement (2550 mg/day).
2000). Androgenic changes such as acne and Treatment usually starts with 25 mg/day. Serum
hirsutism, including facial hair growth, have been DHEAS concentrations can easily be monitored and
reported as possible side-effects in numerous con- should be in the respective sex- and age-adjusted
trolled trials (Arlt et al. 1999b; Hunt et al. 2000; reference range (Orentreich et al. 1984). It is import-
Lovas et al. 2003; Brooke et al. 2006; Gurnell ant to know that significant improvement may occur
et al. 2008). only after two to four months of treatment.
Treatment of elderlies with age-related physiolo-
gically low endogenous DHEA(S) is not justified. All
20.7.5 Immune system available evidence indicates that the age-related
Based on data from animal experiments (Svec and decline in DHEA(S) concentration is not necessarily
Porter 1998) and from in-vitro studies (Meikle et al. associated with impairment in well-being and mood
1992; Okabe et al. 1995), DHEA has been suggested or with increased fatigue. Accordingly, DHEA sup-
as a steroid with immune-regulatory activity. This plementation offers no apparent benefit for such a
view is supported by the clinical studies in patients population. This is a situation very similar to post-
with SLE, demonstrating glucocorticoid-sparing menopausal hormone replacement: despite very low
activity of DHEA and clinical improvement (Chang estradiol concentrations, estrogen replacement may
et al. 2002). However, in these studies DHEA was be more often detrimental than beneficial. This does
given at a clearly supraphysiological dose (200 mg/d), not exclude the possibility that certain subgroups
and physiological replacement doses (50 mg/d) of elderly subjects (e.g. patients with frailty) may
given to healthy elderlies in the DHEAge study benefit from DHEA supplementation, but these sub-
did not have any effect on B- and T-cell popula- groups need to be defined. In particular, there is no
tions, cytokine production or natural killer (NK) evidence that DHEA supplementation reverses rele-
cell cytotoxicity (unpublished observations). In- vant aspects of aging.
vitro studies with human cells also show DHEA- Reports on beneficial effects of DHEA on a dimin-
induced increases in IL-2 secretion (Suzuki et al. ished ovarian reserve in IVF patients are intriguing
1991) and NK cell activity (Solerte et al. 1999) and but still inconclusive. Thus, despite the widespread
inhibition of IL-6 release (Gordon et al. 2001). use of pharmacological doses of DHEA (75 mg/day)
Secretion of IL-2 in SLE correlates with circulating in this setting, its use should be regarded as clearly
DHEAS, and in-vitro DHEA restores IL-2 secretion experimental pending larger-scale, randomized stud-
from T-lymphocytes of SLE patients (Suzuki et al. ies that clearly demonstrate effectiveness.
1995). No consistent in-vivo data on immune
effects of DHEA in humans are reported. Again
it is likely that beneficial effects of DHEA are 20.9 Future perspectives
more easily detectable in patients with immunopa- Important progress has been made in the field of
thies and an altered immune system at baseline DHEA research. The therapeutic potential of DHEA
(Hazeldine et al. 2010). is more clearly visible (see Table 20.1), and DHEA has

449
Chapter 20: Dehydroepiandrosterone and androstenedione

Table 20.1 Action profile of DHEA immunopathies and women with androgen defi-
Central nervous system:
ciency-related complaints (e. g. loss of libido).
In these patient groups administration of DHEA
 Mood ! ("), well-being " (!), anxiety # (!), must not be regarded as substitution therapy but
depression #, fatigue ! (#), cognitive function ! rather as pharmacotherapy. Accordingly, only large,
 Libido ! (" in women), sexual satisfaction ! (") prospective, randomized, double-blind trials will
Metabolism:
allow definition of the benefits and also the risks of
such DHEA pharmacotherapy. Much work remains
 Insulin sensitivity !, fasting glucose ! to be done in this area.
 HDL cholesterol # (!), total cholesterol ! An important contribution to the development of
 IGF-1 " (!)
treatment strategies with DHEA will come from a
better understanding of the mechanisms of action of
Muscle: DHEA. It is predicted that future DHEA research
 Strength ! ("), muscle area ! will be successful in identifying more specific
 Lean body mass ! (")
direct actions of DHEA; e.g. the identification and
characterization of the putative membrane-bound
Bone: G-protein-coupled DHEA receptor. Of particular
 Bone mineral density ! (" in women) interest will be the investigation of specific DHEA
 Bone maker ! ("#)
actions on the immune and central nervous systems.
In conclusion, DHEA has emerged as a fascinating
Skin: adrenal steroid, but its physiology and therapeutic
 Sebum production " potential are still waiting to be fully revealed.
 Skin hydration "
20.10 Key messages
 Acne "
 DHEA(S) secretion shows a characteristic pattern
Immune system: during the human life cycle, with a prepubertal
 Glucocorticoid demand in immunopathies (#) rise (adrenarche) and a continuous decline
(adrenopause) after a peak in early adulthood.
 Immune cell distribution ! (B-cells, T-cells, NK cells) There is high interindividual variability and a sex
Symbols: " increase; ! unchanged; # reduced; () changes only difference in circulating DHEA(S) levels with
found in some of the published studies. higher concentrations in males.
 Low DHEA(S) concentrations predict imminent
mortality in both men and women. Low DHEAS
become part of routine replacement in many patients levels may be a non-specific marker of poor
with adrenal insufficiency and impaired well-being. health and rather an epiphenomenon but not a
cause of disease.
However, large phase III trials are still necessary to
 Dehydroepiandrosterone exerts its biological
firmly establish its role in the treatment of adrenal activity via its downstream conversion to
failure and to further define the spectrum of DHEA potent sex steroids, as a neurosteroid interacting
actions. In particular, it is difficult to separate andro- with various receptors in the central nervous
genic from DHEA-specific effects. In this regard stud- system, and also via a membrane-bound specific
ies in male patients with adrenal insufficiency will G-protein-coupled DHEA receptor, particularly
be highly informative, as in this population DHEA- in the vascular system.
induced androgenic actions are expected to play  Like DHEA, androstenedione acts as a
virtually no role on the background of high endogen- prohormone and is converted predominantly
ous testosterone concentrations. into androgens and to a minor extent into
Recent larger trials have again demonstrated estrogens after oral administration.
 Treatment of patients with adrenal
that hopes of using DHEA as an anti-aging remedy
insufficiency (2550 mg/day DHEA) improves
were not justified. However, DHEA may have QoL, depression and fatigue and may also
therapeutic potential for patients with psychiatric ill- improve sexuality in female patients. By
nesses (depression, schizophrenia, dysthymia) or

450
Chapter 20: Dehydroepiandrosterone and androstenedione

contrast, in healthy elderly subjects with


age-related low endogenous DHEA(S), beneficial
effects of DHEA supplementation remain
doubtful.
 Dehydroepiandrosterone administration has
been found to improve anxiety and depression in
midlife dysthymia, patients with depression
and schizophrenia. In systemic lupus
erythematous, DHEA reduces disease activity
and flares, and allows reduction of the
glucocorticoid dose.
 In short-term studies, androstenedione and DHEA
did not improve muscle function or muscle
strength, although these steroids are prohibited
as ergogenic drugs by the World Anti Doping
Agency (WADA).
 The available clinical evidence suggests that the
main target tissues for DHEA are the central
nervous system, the skin and possibly the
immune system. Beneficial effects on muscle
function, bone and ovarian folliculogenesis
remain to be fully established.

20.11 References to dehydroepiandrosterone: a pilot


study. Aliment Pharmacol Ther
Dehydroepiandrosterone
replacement in women with adrenal
Achermann JC, Silverman BL (2001) 17:409414 insufficiency. N Engl J Med
Dehydroepiandrosterone 341:10131020
replacement for patients with Arlt W (2003) Management of the
adrenal insufficiency. Lancet androgen-deficient woman. Growth Arlt W, Callies F, Allolio B (2000)
357:13811382 Horm IGF Res 13 (Suppl A): S85S89 DHEA replacement in women with
Arlt W, Justl HG, Callies F, Reincke M, adrenal insufficiency
Alkatib AA, Cosma M, Elamin MB,
Hubler D, Oettel M, Ernst M, pharmacokinetics, bioconversion
Erickson D, Swiglo BA, Erwin PJ,
Schulte HM, Allolio B (1998) Oral and clinical effects on well-being,
Montori VM (2009) A systematic
dehydroepiandrosterone for adrenal sexuality and cognition. Endocr Res
review and meta-analysis of
androgen replacement: 26:505511
randomized placebo-controlled
trials of DHEA treatment effects on pharmacokinetics and peripheral Arlt W, Callies F, Koehler I, van
quality of life in women with conversion to androgens and Vlijmen JC, Fassnacht M,
adrenal insufficiency. estrogens in young healthy females Strasburger CJ, Seibel MJ,
J Clin Endocrinol Metab after dexamethasone suppression. Huebler D, Ernst M, Oettel M,
94:36763681 J Clin Endocrinol Metab Reincke M, Schulte HM, Allolio B
83:19281934 (2001) Dehydroepiandrosterone
Allolio B, Arlt W (2002) DHEA
Arlt W, Haas J, Callies F, Reincke M, supplementation in healthy men
treatment: myth or reality? Trends
with an age-related decline of
Endocrinol Metab 13:288294 Hubler D, Oettel M, Ernst M, Schulte
HM, Allolio B (1999a) dehydroepiandrosterone secretion.
Allolio B, Arlt W, Hahner S (2007) Biotransformation of oral J Clin Endocrinol Metab
DHEA: why, when, and how dehydroepiandrosterone in elderly 86:46864692
much DHEA replacement in men: significant increase in Arlt W, Hammer F, Sanning P, Butcher
adrenal insufficiency. Ann circulating estrogens. J Clin SK, Lord JM, Allolio B, Annane D,
Endocrinol (Paris) 68:268273 Endocrinol Metab 84:21702176 Stewart PM (2006) Dissociation of
Andus T, Klebl F, Rogler G, Bregenzer N, serum dehydroepiandrosterone and
Arlt W, Callies F, van Vlijmen JC,
Scholmerich J, Straub RH (2003) dehydroepiandrosterone sulfate in
Koehler I, Reincke M, Bidlingmaier M,
Patients with refractory Crohns septic shock. J Clin Endocrinol
Huebler D, Oettel M, Ernst M,
disease or ulcerative colitis respond
Schulte HM, Allolio B (1999b) Metab 91:25482554

451
Chapter 20: Dehydroepiandrosterone and androstenedione

Auchus RJ, Rainey WE (2004) Dehydroepiandrosterone (DHEA), hypopituitarism. J Pediatr


Adrenarche physiology, DHEA sulfate, and aging: Endocrinol Metab 18:355362
biochemistry and human disease. contribution of the DHEAge study Binder G, Weber S, Ehrismann M,
Clin Endocrinol (Oxf) 60:288296 to a sociobiomedical issue. Proc Natl Zaiser N, Meisner C, Ranke MB,
Barad D, Brill H, Gleicher N (2007) Acad Sci USA 97:42794284 Maier L, Wudy SA, Hartmann MF,
Update on the use of Beckham SG, Earnest CP (2003) Four Heinrich U, Bettendorf M, Doerr
dehydroepiandrosterone weeks of androstenedione HG, Pfaeffle RW, Keller E (2009)
supplementation among women supplementation diminishes the Effects of dehydroepiandrosterone
with diminished ovarian function. treatment response in middle therapy on pubic hair growth and
J Assist Reprod Genet 24:629634 aged men. Br J Sports Med psychological well-being in
Barnhart KT, Freeman E, Grisso JA, 37:212218 adolescent girls and young women
Rader DJ, Sammel M, Kapoor S, with central adrenal insufficiency:
Bensing S, Brandt L, Tabaroj F, Sjoberg O,
Nestler JE (1999) The effect of a double-blind, randomized, placebo-
Nilsson B, Ekbom A, Blomqvist P,
dehydroepiandrosterone controlled phase III trial. J Clin
Kampe O (2008) Increased death
supplementation to symptomatic Endocrinol Metab 94:11821190
risk and altered cancer incidence
perimenopausal women on serum pattern in patients with isolated or Bloch M, Schmidt PJ, Danaceau MA,
endocrine profiles, lipid parameters, combined autoimmune primary Adams LF, Rubinow DR (1999)
and health-related quality of life. adrenocortical insufficiency. Clin Dehydroepiandrosterone treatment
J Clin Endocrinol Metab Endocrinol (Oxf) 69:697704 of midlife dysthymia. Biol
84:38963902 Psychiatry 45:15331541
Bergthorsdottir R, Leonsson-
Barrett-Connor E, Goodman-Gruen D Zachrisson M, Oden A, Johannsson Brooke AM, Kalingag LA, Miraki-
(1995) Dehydroepiandrosterone G (2006) Premature mortality in Moud F, Camacho-Hubner C,
sulfate does not predict patients with Addisons disease: a Maher KT, Walker DM, Hinson JP,
cardiovascular death in population-based study. Monson JP (2006)
postmenopausal women. The J Clin Endocrinol Metab Dehydroepiandrosterone improves
Rancho Bernardo Study. Circulation 91:48494853 psychological well-being in male
91:17571760 and female hypopituitary patients
Berr C, Lafont S, Debuire B, Dartigues on maintenance growth hormone
Bassindale T, Cowan DA, Dale S, Hutt JF, Baulieu EE (1996) Relationships
AJ, Leeds AR, Wheeler MJ, Kicman replacement. J Clin Endocrinol
of dehydroepiandrosterone sulfate Metab 91:37733779
AT (2004) Effects of oral in the elderly with functional,
administration of androstenedione psychological, and mental status, Brown GA, Vukovich MD, Martini ER,
on plasma androgens in young and short-term mortality: a Kohut ML, Franke WD, Jackson
women using hormonal French community-based study. DA, King DS (2000) Endocrine
contraception. J Clin Endocrinol Proc Natl Acad Sci USA responses to chronic
Metab 89:60306038 93:1341013415 androstenedione intake in 30- to
56-year-old men. J Clin Endocrinol
Basu R, Dalla Man C, Campioni M, Bhuiyan MS, Tagashira H, Fukunaga K Metab 85:40744080
Basu A, Nair KS, Jensen MD, (2011) Dehydroepiandrosterone-
Khosla S, Klee G, Toffolo G, mediated stimulation of sigma-1 Brown GA, Martini ER, Roberts BS,
Cobelli C, Rizza RA (2007) Two receptor activates Akt-eNOS Vukovich MD, King DS (2002)
years of treatment with signaling in the thoracic aorta of Acute hormonal response to
dehydroepiandrosterone does not ovariectomized rats with abdominal sublingual androstenediol intake
improve insulin secretion, insulin aortic banding. Cardiovasc Ther in young men. J Appl Physiol
action, or postprandial glucose 29:219230 92:142146
turnover in elderly men or women. Brown GA, Dewey JC, Brunkhorst JA,
Bhuiyan S, Fukunaga K (2010)
Diabetes 56:753766 Vukovich MD, King DS (2004)
Stimulation of Sigma-1 receptor by
Baulieu EE, Thomas G, Legrain S, dehydroepiandrosterone Changes in serum testosterone and
Lahlou N, Roger M, Debuire B, ameliorates hypertension-induced estradiol concentrations following
Faucounau V, Girard L, Hervy MP, kidney hypertrophy in acute androstenedione ingestion in
Latour F, Leaud MC, Mokrane A, ovariectomized rats. Exp Biol Med young women. Horm Metab Res
Pitti-Ferrandi H, Trivalle C, de (Maywood) 235:356364 36:6266
Lacharriere O, Nouveau S, Rakoto- Bilger M, Speraw S, LaFranchi SH, Calfee R, Fadale P (2006) Popular
Arison B, Souberbielle JC, Raison J, Hanna CE (2005) Androgen ergogenic drugs and supplements
Le Bouc Y, Raynaud A, Girerd X, replacement in adolescents and in young athletes. Pediatrics
Forette F (2000) young women with 117:e577e589

452
Chapter 20: Dehydroepiandrosterone and androstenedione

Callies F, Fassnacht M, van Vlijmen JC, replacement for postmenopausal Giagulli VA, Verdonck L, Giorgino R,
Koehler I, Huebler D, Seibel MJ, women. J Clin Endocrinol Metab Vermeulen A (1989) Precursors of
Arlt W, Allolio B (2001) 96:16421653 plasma androstanediol- and
Dehydroepiandrosterone Dayal M, Sammel MD, Zhao J, androgen-glucuronides in women.
replacement in women with adrenal Hummel AC, Vandenbourne K, J Steroid Biochem 33:935940
insufficiency: effects on body Barnhart KT (2005) Gleicher N, Barad DH (2011)
composition, serum leptin, bone Supplementation with DHEA: effect Dehydroepiandrosterone (DHEA)
turnover, and exercise capacity. on muscle size, strength, quality of supplementation in diminished
J Clin Endocrinol Metab life, and lipids. J Womens Health ovarian reserve (DOR). Reprod Biol
86:19681972 (Larchmt) 14:391400 Endocrinol 9: 67
Casson PR, Faquin LC, Stentz FB, Deighton CM, Watson MJ, Walker DJ Gleicher N, Weghofer A, Barad DH
Straughn AB, Andersen RN, (1992) Sex hormones in (2010) Dehydroepiandrosterone
Abraham GE, Buster JE (1995) postmenopausal HLA-identical (DHEA) reduces embryo aneuploidy:
Replacement of rheumatoid arthritis discordant direct evidence from preimplantation
dehydroepiandrosterone enhances sibling pairs. J Rheumatol genetic screening (PGS). Reprod Biol
T-lymphocyte insulin binding in 19:16631667 Endocrinol 8:140
postmenopausal women. Fertil Steril
63:10271031 Dhatariya K, Bigelow ML, Nair KS Gordon CM, LeBoff MS, Glowacki J
(2005) Effect of (2001) Adrenal and gonadal steroids
Casson PR, Lindsay MS, Pisarska MD, dehydroepiandrosterone inhibit IL-6 secretion by human
Carson SA, Buster JE (2000) replacement on insulin sensitivity marrow cells. Cytokine 16:178186
Dehydroepiandrosterone and lipids in hypoadrenal women.
supplementation augments ovarian Gurnell EM, Hunt PJ, Curran SE,
Diabetes 54:765769
stimulation in poor responders: Conway CL, Pullenayegum EM,
a case series. Hum Reprod Dhatariya KK, Greenlund LJ, Bigelow Huppert FA, Compston JE, Herbert
15:21292132 ML, Thapa P, Oberg AL, Ford GC, J, Chatterjee VK (2008) Long-term
Schimke JM, Nair KS (2008) DHEA replacement in primary
Chang DM, Lan JL, Lin HY, Luo SF Dehydroepiandrosterone adrenal insufficiency: a randomized,
(2002) Dehydroepiandrosterone replacement therapy in hypoadrenal controlled trial. J Clin Endocrinol
treatment of women with mild-to- women: protein anabolism and Metab 93:400409
moderate systemic lupus skeletal muscle function. Mayo Clin
erythematosus: a multicenter Hackbert L, Heiman JR (2002) Acute
Proc 83:12181225
randomized, double-blind, placebo- dehydroepiandrosterone (DHEA)
controlled trial. Arthritis Rheum Diamond P, Cusan L, Gomez JL, effects on sexual arousal in
46:29242927 Belanger A, Labrie F (1996) postmenopausal women. J Womens
Metabolic effects of 12-month Health Gend Based Med 11:155162
Compagnone NA, Mellon SH (1998) percutaneous
Dehydroepiandrosterone: a Hahner S, Loeffler M, Fassnacht M,
dehydroepiandrosterone
potential signalling molecule for Weismann D, Koschker AC,
replacement therapy in
neocortical organization during Quinkler M, Decker O, Arlt W,
postmenopausal women.
development. Proc Natl Acad Sci Allolio B (2007) Impaired subjective
J Endocrinol 150 Suppl: S43S50
USA 95:46784683 health status in 256 patients with
Erichsen MM, Lovas K, Skinningsrud B, adrenal insufficiency on standard
Cumming DC, Rebar RW, Hopper BR, Wolff AB, Undlien DE, Svartberg J, therapy based on cross-sectional
Yen SS (1982) Evidence for an Fougner KJ, Berg TJ, Bollerslev J, analysis. J Clin Endocrinol Metab
influence of the ovary on circulating Mella B, Carlson JA, Erlich H, 92:39123922
dehydroepiandrosterone sulfate Husebye ES (2009) Clinical,
levels. J Clin Endocrinol Metab immunological, and genetic Hammer F, Subtil S, Lux P, Maser-
54:10691071 features of autoimmune primary
Gluth C, Stewart PM, Allolio B, Arlt
adrenal insufficiency: W (2005) No evidence for hepatic
Danenberg HD, Ben-Yehuda A, Zakay-
conversion of
Rones Z, Gross DJ, Friedman G observations from a Norwegian
registry. J Clin Endocrinol Metab dehydroepiandrosterone (DHEA)
(1997) Dehydroepiandrosterone
sulfate to DHEA: in vivo and in
treatment is not beneficial to the 94:48824890
vitro studies. J Clin Endocrinol
immune response to influenza in Flynn MA, Weaver-Osterholtz D, Metab 90:36003605
elderly subjects. J Clin Endocrinol Sharpe-Timms KL, Allen S, Krause
Metab 82:29112914 G (1999) Dehydroepiandrosterone Hartkamp A, Geenen R, Godaert GL,
replacement in aging humans. J Clin Bijl M, Bijlsma JW, Derksen RH
Davis SR, Panjari M, Stanczyk FZ
Endocrinol Metab 84:15271533 (2010) Effects of
(2011) Clinical review: DHEA

453
Chapter 20: Dehydroepiandrosterone and androstenedione

dehydroepiandrosterone on fatigue J Clin Endocrinol Metab Labrie F (2010) DHEA, important


and well-being in women with 93:47674773 source of sex steroids in men and
quiescent systemic lupus Johannsson G, Burman P, Wiren L, even more in women. Prog Brain
erythematosus: a randomised Engstrom BE, Nilsson AG, Res 182:97148
controlled trial. Ann Rheum Dis Ottosson M, Jonsson B, Bengtsson BA, Labrie F, Diamond P, Cusan L, Gomez
69:11441147 Karlsson FA (2002) Low dose JL, Belanger A, Candas B (1997)
Hazeldine J, Arlt W, Lord JM (2010) dehydroepiandrosterone affects Effect of 12-month
Dehydroepiandrosterone as a behavior in hypopituitary dehydroepiandrosterone
regulator of immune cell function. androgen-deficient women: a replacement therapy on bone,
J Steroid Biochem Mol Biol placebo-controlled trial. J Clin vagina, and endometrium in
120:127136 Endocrinol Metab 87:20462052 postmenopausal women.
J Clin Endocrinol Metab
Hunt PJ, Gurnell EM, Huppert FA, Kahn AJ, Halloran B, Wolkowitz O,
82:34983505
Richards C, Prevost AT, Wass JA, Brizendine L (2002)
Herbert J, Chatterjee VK (2000) Dehydroepiandrosterone Labrie F, Luu-The V, Labrie C,
Improvement in mood and fatigue supplementation and bone turnover Belanger A, Simard J, Lin SX,
after dehydroepiandrosterone in middle-aged to elderly men. J Clin Pelletier G (2003) Endocrine and
replacement in Addisons disease in a Endocrinol Metab 87:15441549 intracrine sources of androgens in
randomized, double blind trial. J Clin women: inhibition of breast cancer
Kicman AT, Bassindale T, Cowan DA,
Endocrinol Metab 85:46504656 and other roles of androgens and
Dale S, Hutt AJ, Leeds AR (2003)
their precursor
Igwebuike A, Irving BA, Bigelow ML, Effect of androstenedione ingestion
dehydroepiandrosterone. Endocr
Short KR, McConnell JP, on plasma testosterone in young
Rev 24:152182
Nair KS (2008) Lack of women; a dietary supplement with
dehydroepiandrosterone effect on a potential health risks. Clin Chem Labrie F, Archer D, Bouchard C,
combined endurance and resistance 49:167169 Fortier M, Cusan L, Gomez JL,
exercise program in Girard G, Baron M, Ayotte N,
Kim SS, Brody KH (2001)
postmenopausal women. J Clin Moreau M, Dube R, Cote I,
Dehydroepiandrosterone
Endocrinol Metab 93:534538 Labrie C, Lavoie L, Berger L, Gilbert
replacement in Addisons disease.
L, Martel C, Balser J (2009) Effect of
Iruthayanathan M, OLeary B, Paul G, Eur J Obstet Gynecol Reprod Biol
intravaginal
Dillon JS (2011) Hydrogen peroxide 97:9697
dehydroepiandrosterone
signaling mediates DHEA-induced King DS, Sharp RL, Vukovich MD, (Prasterone) on libido and sexual
vascular endothelial cell Brown GA, Reifenrath TA, Uhl NL, dysfunction in postmenopausal
proliferation. Steroids 76:14831490 Parsons KA (1999) Effect of oral women. Menopause 16:923931
Jakobi JM, Killinger DW, Wolfe BM, androstenedione on serum Lahita RG, Bradlow HL, Ginzler E,
Mahon JL, Rice CL (2001) testosterone and adaptations to
Pang S, New M (1987) Low plasma
Quadriceps muscle function and resistance training in young men: androgens in women with systemic
fatigue in women with Addisons a randomized controlled trial.
lupus erythematosus. Arthritis
disease. Muscle Nerve 24:10401049 JAMA 281:20202028 Rheum 30:241248
Jankowski CM, Gozansky WS, Kline MD, Jaggers ED (1999) Mania
Laughlin GA, Barrett-Connor E (2000)
Schwartz RS, Dahl DJ, Kittelson JM, onset while using
Sexual dimorphism in the influence
Scott SM, Van Pelt RE, Kohrt WM dehydroepiandrosterone.
of advanced aging on adrenal
(2006) Effects of Am J Psychiatry 156: 971
hormone levels: the Rancho
dehydroepiandrosterone Kolibianakis EM, Venetis CA, Tarlatzis Bernardo Study. J Clin Endocrinol
replacement therapy on bone BC (2011) DHEA administration in Metab 85:35613568
mineral density in older adults: a poor responders. Hum Reprod
Laurine E, Lafitte D, Gregoire C,
randomized, controlled trial. J Clin 26:730731; author reply:731
Endocrinol Metab 91:29862993 Seree E, Loret E, Douillard S,
Kritz-Silverstein D, von Muhlen D, Michel B, Briand C, Verdier JM
Jankowski CM, Gozansky WS, Laughlin GA, Bettencourt R (2008) (2003) Specific binding of
Kittelson JM, Van Pelt RE, Schwartz Effects of dehydroepiandrosterone dehydroepiandrosterone to the
RS, Kohrt WM (2008) Increases in supplementation on cognitive N terminus of the microtubule-
bone mineral density in response to function and quality of life: the associated protein MAP2. J Biol
oral dehydroepiandrosterone DHEA and Well-Ness (DAWN) Chem 278:2997929986
replacement in older adults appear Trial. J Am Geriatr Soc
to be mediated by serum estrogens. 56:12921298 Lazaridis I, Charalampopoulos I,
Alexaki VI, Avlonitis N,

454
Chapter 20: Dehydroepiandrosterone and androstenedione

Pediaditakis I, Efstathopoulos P, Dehydroepiandrosterone stimulates dehydroepiandrosterone (DHEA)


Calogeropoulou T, Castanas E, endothelial proliferation and on circulating sex steroids, body
Gravanis A (2011) Neurosteroid angiogenesis through extracellular composition and muscle strength in
dehydroepiandrosterone interacts signal-regulated kinase 1/2- age-advanced men and women.
with nerve growth factor (NGF) mediated mechanisms. Clin Endocrinol (Oxf) 49:421432
receptors, preventing Endocrinology 149:889898 Nair KS, Rizza RA, OBrien P,
neuronal apoptosis. PLoS Biol 9: Lovas K, Gebre-Medhin G, Trovik TS, Dhatariya K, Short KR, Nehra A,
e1001051 Fougner KJ, Uhlving S, Nedrebo Vittone JL, Klee GG, Basu A,
Leder BZ, Longcope C, Catlin DH, BG, Myking OL, Kampe O, Basu R, Cobelli C, Toffolo G,
Ahrens B, Schoenfeld DA, Husebye ES (2003) Replacement of Dalla Man C, Tindall DJ, Melton LJ
Finkelstein JS (2000) Oral dehydroepiandrosterone in adrenal 3rd, Smith GE, Khosla S, Jensen MD
androstenedione administration failure: no benefit for subjective (2006) DHEA in elderly women and
and serum testosterone health status and sexuality in a DHEA or testosterone in
concentrations in young men. 9-month, randomized, parallel elderly men. N Engl J Med
JAMA 283:779782 group clinical trial. J Clin Endocrinol 355:16471659
Metab 88:11121118 Nieschlag E, Loriaux DL, Ruder HJ,
Leder BZ, Catlin DH, Longcope C,
Ahrens B, Schoenfeld DA, Finkelstein Markowitz JS, Carson WH, Jackson Zucker IR, Kirschner MA, Lipsett
JS (2001) Metabolism of orally CW (1999) Possible MB (1973) The secretion of
administered androstenedione in dihydroepiandrosterone- dehydroepiandrosterone and
young men. J Clin Endocrinol Metab induced mania. Biol Psychiatry dehydroepiandrosterone sulphate in
86:36543658 45:241242 man. J Endocrinol 57:123134
Leder BZ, Leblanc KM, Longcope C, Mazat L, Lafont S, Berr C, Debuire B, Noordam C, Dhir V, McNelis JC,
Lee H, Catlin DH, Finkelstein JS Tessier JF, Dartigues JF, Baulieu EE Schlereth F, Hanley NA, Krone N,
(2002) Effects of oral (2001) Prospective measurements of Smeitink JA, Smeets R, Sweep FC,
androstenedione administration on dehydroepiandrosterone sulfate in a Claahsen-van der Grinten HL, Arlt
serum testosterone and estradiol cohort of elderly subjects: W (2009) Inactivating PAPSS2
levels in postmenopausal women. relationship to gender, subjective mutations in a patient with
J Clin Endocrinol Metab health, smoking habits, and 10-year premature pubarche. N Engl J Med
87:54495454 mortality. Proc Natl Acad Sci USA 360:23102318
98:81458150 Okabe T, Haji M, Takayanagi R,
Libe R, Barbetta L, DallAsta C,
Salvaggio F, Gala C, Beck-Peccoz P, Meikle AW, Dorchuck RW, Araneo Adachi M, Imasaki K, Kurimoto F,
Ambrosi B (2004) Effects of BA, Stringham JD, Evans TG, Watanabe T, Nawata H (1995)
dehydroepiandrosterone (DHEA) Spruance SL, Daynes RA (1992) The Up-regulation of high-affinity
supplementation on hormonal, presence of a dehydroepiandrosterone binding
metabolic and behavioral status in dehydroepiandrosterone-specific activity by dehydroepiandrosterone
patients with hypoadrenalism. receptor binding complex in in activated human T lymphocytes.
J Endocrinol Invest 27: murine T cells. J Steroid Biochem J Clin Endocrinol Metab
736741 Mol Biol 42:293304 80:29932996
Lindschau C, Kirsch T, Klinge U, Melez KA, Boegel WA, Steinberg AD Orentreich N, Brind JL, Rizer RL,
Kolkhof P, Peters I, Fiebeler A (1980) Therapeutic studies in New Vogelman JH (1984) Age changes
(2011) Dehydroepiandrosterone- Zealand mice. VII. Successful and sex differences in serum
induced phosphorylation and androgen treatment of NZB/NZW dehydroepiandrosterone sulfate
translocation of FoxO1 depend on F1 females of different ages. concentrations throughout
the mineralocorticoid receptor. Arthritis Rheum 23:4147 adulthood. J Clin Endocrinol Metab
Hypertension 58:471478 Morales AJ, Nolan JJ, Nelson JC, Yen 59:551555
Liu D, Dillon JS (2002) SS (1994) Effects of replacement Orentreich N, Brind JL, Vogelman JH,
Dehydroepiandrosterone activates dose of dehydroepiandrosterone in Andres R, Baldwin H (1992) Long-
endothelial cell nitric-oxide men and women of advancing term longitudinal measurements of
synthase by a specific plasma age. J Clin Endocrinol Metab plasma dehydroepiandrosterone
membrane receptor coupled to 78:13601367 sulfate in normal men. J Clin
Galpha(i2,3). J Biol Chem Morales AJ, Haubrich RH, Hwang JY, Endocrinol Metab 75:10021004
277:2137921388 Asakura H, Yen SS (1998) The effect Panjari M, Bell RJ, Jane F, Wolfe R,
Liu D, Iruthayanathan M, Homan LL, of six months treatment with a 100 Adams J, Morrow C, Davis SR
Wang Y, Yang L, Dillon JS (2008) mg daily dose of (2009) A randomized trial of oral

455
Chapter 20: Dehydroepiandrosterone and androstenedione

DHEA treatment for sexual randomized, placebo-controlled constitutionally delayed growth and
function, well-being, and study. Urology 53:590594; adolescence. J Clin Endocrinol
menopausal symptoms in discussion:594595 Metab 51:548556
postmenopausal women with low Roth MY, Page ST, Lin K, Anawalt BD, Solerte SB, Fioravanti M, Vignati G,
libido. J Sex Med 6:25792590 Matsumoto AM, Marck B, Bremner Giustina A, Cravello L, Ferrari E
Parker CR Jr, Mixon RL, Brissie RM, WJ, Amory JK (2011) The effect of (1999) Dehydroepiandrosterone
Grizzle WE (1997) Aging alters gonadotropin withdrawal and sulfate enhances natural killer cell
zonation in the adrenal cortex of stimulation with human chorionic cytotoxicity in humans via locally
men. J Clin Endocrinol Metab gonadotropin on intratesticular generated immunoreactive
82:38983901 androstenedione and DHEA in insulin-like growth factor I.
Percheron G, Hogrel JY, Denot- normal men. J Clin Endocrinol J Clin Endocrinol Metab
Ledunois S, Fayet G, Forette F, Metab 96:11751181 84:32603267
Baulieu EE, Fardeau M, Marini JF Schmidt PJ, Daly RC, Bloch M, Smith Spencer JB, Klein M, Kumar A, Azziz R
(2003) Effect of 1-year oral MJ, Danaceau MA, St Clair LS, (2007) The age-associated decline of
administration of Murphy JH, Haq N, Rubinow DR androgens in reproductive age and
dehydroepiandrosterone to 60- to (2005) Dehydroepiandrosterone menopausal Black and White
80-year-old individuals on muscle monotherapy in midlife-onset women. J Clin Endocrinol Metab
function and cross-sectional area: a major and minor depression. 92:47304733
double-blind placebo-controlled Arch Gen Psychiatry 62:154162 Srinivasan M, Irving BA, Frye RL,
trial. Arch Intern Med 163:720727 OBrien P, Hartman SJ, McConnell
Sen A, Hammes SR (2010) Granulosa
Perez-Neri I, Montes S, Ojeda-Lopez C, cell-specific androgen receptors are JP, Nair KS (2010) Effects on
Ramirez-Bermudez J, Rios C (2008) critical regulators of ovarian lipoprotein particles of long-term
Modulation of neurotransmitter development and function. Mol dehydroepiandrosterone in elderly
systems by dehydroepiandrosterone Endocrinol 24:13931403 men and women and testosterone in
and dehydroepiandrosterone elderly men. J Clin Endocrinol
Shifren JL, Braunstein GD, Simon JA,
sulfate: mechanism of action and Metab 95:16171625
Casson PR, Buster JE, Redmond
relevance to psychiatric disorders. Stanway SJ, Delavault P, Purohit A,
GP, Burki RE, Ginsburg ES, Rosen
Prog Neuropsychopharmacol Biol Woo LW, Thurieau C, Potter BV,
RC, Leiblum SR, Caramelli KE,
Psychiatry 32:11181130 Reed MJ (2007) Steroid sulfatase: a
Mazer NA (2000) Transdermal
Radford DJ, Wang K, McNelis JC, testosterone treatment in women new target for the endocrine therapy
Taylor AE, Hechenberger G, with impaired sexual function after of breast cancer. Oncologist
Hofmann J, Chahal H, Arlt W, Lord oophorectomy. N Engl J Med 12:370374
JM (2010) Dehdyroepiandrosterone 343:682688 Stickney DR, Ahlem CN, Morgan E,
sulfate directly activates protein Reading CL, Onizuka N, Frincke JM
Shufelt C, Bretsky P, Almeida CM,
kinase C-beta to increase human (2011) Phase I and phase II clinical
Johnson BD, Shaw LJ, Azziz R,
neutrophil superoxide trials of androst-5-ene-
Braunstein GD, Pepine CJ,
generation. Mol Endocrinol 3beta,7beta,17beta-triol. Am
Bittner V, Vido DA, Stanczyk FZ,
24:813821 J Transl Res 3:275283
Bairey Merz CN (2010) DHEA-S
Rasmussen BB, Volpi E, Gore DC, levels and cardiovascular disease Strous RD, Maayan R, Lapidus R,
Wolfe RR (2000) Androstenedione mortality in postmenopausal Stryjer R, Lustig M, Kotler M,
does not stimulate muscle protein women: results from the National Weizman A (2003)
anabolism in young healthy men. Institutes of Health National Dehydroepiandrosterone
J Clin Endocrinol Metab 85:5559 Heart, Lung, and Blood Institute augmentation in the management
Reed MJ, Purohit A, Woo LW, (NHLBI)-sponsored Womens of negative, depressive, and
Newman SP, Potter BV (2005) Ischemia Syndrome Evaluation anxiety symptoms in schizophrenia.
Steroid sulfatase: molecular biology, (WISE). J Clin Endocrinol Metab Arch Gen Psychiatry 60:133141
regulation, and inhibition. Endocr 95:49854992
Sugino M, Ohsawa N, Ito T, Ishida S,
Rev 26:171202 Sklar CA, Kaplan SL, Grumbach MM Yamasaki H, Kimura F, Shinoda K
Reiter WJ, Pycha A, Schatzl G, (1980) Evidence for dissociation (1998) A pilot study of
Pokorny A, Gruber DM, Huber JC, between adrenarche and dehydroepiandrosterone sulfate in
Marberger M (1999) gonadarche: studies in patients with myotonic dystrophy. Neurology
Dehydroepiandrosterone in the idiopathic precocious puberty, 51:586589
treatment of erectile dysfunction: a gonadal dysgenesis, isolated
prospective, double-blind, gonadotropin deficiency, and

456
Chapter 20: Dehydroepiandrosterone and androstenedione

Sunkara SK, Coomarasamy A (2011) Psychoneuroendocrinology hormones in human behavior.


Androgen pretreatment in poor 26:591612 I. Changes in female sexuality after
responders undergoing controlled van Thiel SW, Romijn JA, Pereira AM, adrenalectomy. J Clin Endocrinol
ovarian stimulation and in Biermasz NR, Roelfsema F, van Metab 19:193202
vitro fertilization treatment. Hemert A, Ballieux B, Smit JW Weiss EP, Shah K, Fontana L, Lambert
Fertil Steril 95:e73e74; author (2005) Effects of CP, Holloszy JO, Villareal DT
reply:e75 dehydroepiandrostenedione, (2009) Dehydroepiandrosterone
Suzuki T, Suzuki N, Daynes RA, superimposed on GH substitution, replacement therapy in older adults:
Engleman EG (1991) on quality of life and insulin-like 1- and 2-y effects on bone. Am
Dehydroepiandrosterone enhances growth factor I in patients with J Clin Nutr 89:14591467
IL2 production and cytotoxic secondary adrenal insufficiency: a Williams MR, Ling S, Dawood T,
effector function of human T cells. randomized, placebo-controlled, Hashimura K, Dai A, Li H, Liu JP,
Clin Immunol Immunopathol cross-over trial. J Clin Endocrinol Funder JW, Sudhir K, Komesaroff
61:202211 Metab 90:32953303 PA (2002) Dehydroepiandrosterone
Suzuki T, Suzuki N, Engleman EG, van Vollenhoven RF, Engleman EG, inhibits human vascular smooth
Mizushima Y, Sakane T (1995) Low McGuire JL (1994) An open study muscle cell proliferation
serum levels of of dehydroepiandrosterone in independent of ARs and ERs. J Clin
dehydroepiandrosterone may cause systemic lupus erythematosus. Endocrinol Metab 87:176181
deficient IL-2 production by Arthritis Rheum 37:13051310 Wiser A, Gonen O, Ghetler Y, Shavit T,
lymphocytes in patients with van Vollenhoven RF, Park JL, Berkovitz A, Shulman A
systemic lupus erythematosus Genovese MC, West JP, McGuire JL (2010) Addition of
(SLE). Clin Exp Immunol (1999) A double-blind, placebo- dehydroepiandrosterone (DHEA)
99:251255 controlled, clinical trial of for poor-responder patients before
Svec F, Porter JR (1998) The actions of dehydroepiandrosterone in severe and during IVF treatment improves
exogenous dehydroepiandrosterone systemic lupus erythematosus. the pregnancy rate: a randomized
in experimental animals and Lupus 8:181187 prospective study. Hum Reprod
humans. Proc Soc Exp Biol Med Villareal DT, Holloszy JO (2004) Effect 25:24962500
218:174191 of DHEA on abdominal fat and Wit JM, Langenhorst VJ, Jansen M,
Tomlinson JW, Holden N, Hills RK, insulin action in elderly women and Oostdijk WA, van Doorn J (2001)
Wheatley K, Clayton RN, Bates AS, men: a randomized controlled trial. Dehydroepiandrosterone sulfate
Sheppard MC, Stewart PM (2001) JAMA 292:22432248 treatment for atrichia pubis. Horm
Association between premature Villareal DT, Holloszy JO (2006) Res 56:134139
mortality and hypopituitarism. DHEA enhances effects of weight Wolf OT, Neumann O, Hellhammer
West Midlands Prospective training on muscle mass and DH, Geiben AC, Strasburger CJ,
Hypopituitary Study Group. Lancet strength in elderly women and men. Dressendorfer RA, Pirke KM,
357:425431 Am J Physiol Endocrinol Metab 291: Kirschbaum C (1997) Effects of a
Topor LS, Asai M, Dunn J, Majzoub JA E1003E1008 two-week physiological
(2011) Cortisol stimulates secretion Villareal DT, Holloszy JO, Kohrt WM dehydroepiandrosterone
of dehydroepiandrosterone in (2000) Effects of DHEA substitution on cognitive
human adrenocortical cells through replacement on bone mineral performance and well-being in
inhibition of 3betaHSD2. J Clin density and body composition in healthy elderly women and men.
Endocrinol Metab 96:E31E39 elderly women and men. Clin J Clin Endocrinol Metab
Endocrinol (Oxf) 53:561568 82:23632367
Trivedi DP, Khaw KT (2001)
Dehydroepiandrosterone sulfate von Muhlen D, Laughlin GA, Kritz- Wolf OT, Naumann E, Hellhammer
and mortality in elderly men and Silverstein D, Bergstrom J, DH, Kirschbaum C (1998) Effects of
women. J Clin Endocrinol Metab Bettencourt R (2008) Effect of dehydroepiandrosterone
86:41714177 dehydroepiandrosterone replacement in elderly men on
supplementation on bone mineral event-related potentials, memory,
van Niekerk JK, Huppert FA, Herbert J and well-being. J Gerontol A Biol Sci
(2001) Salivary cortisol and DHEA: density, bone markers, and body
composition in older adults: the Med Sci 53:M385390
association with measures of
cognition and well-being in normal DAWN trial. Osteoporos Int Wolkowitz OM, Reus VI, Keebler A,
older men, and effects of three 19:699707 Nelson N, Friedland M, Brizendine L,
months of DHEA supplementation. Waxenberg SE, Drellich MG, Roberts E (1999) Double-blind
Sutherland AM (1959) The role of treatment of major depression with

457
Chapter 20: Dehydroepiandrosterone and androstenedione

dehydroepiandrosterone. Am effects. Ann N Y Acad Sci Nica A, Penninx BW, Vasan RS,
J Psychiatry 156:646649 774:128142 Rivadeneira F, Small KS, Soranzo N,
Wolkowitz OM, Kramer JH, Reus VI, Young J, Couzinet B, Nahoul K, Brailly S, Uitterlinden AG, Volzke H, Wilson
Costa MM, Yaffe K, Walton P, Chanson P, Baulieu EE, Schaison G SG, Xi L, Zhuang WV, Harris TB,
Raskind M, Peskind E, Newhouse P, (1997) Panhypopituitarism as a Murabito JM, Ohlsson C, Murray
Sack D, De Souza E, Sadowsky C, model to study the metabolism of A, de Jong FH, Spector TD,
Roberts E (2003) DHEA treatment dehydroepiandrosterone (DHEA) in Wallaschofski H (2011) Eight
of Alzheimers disease: a humans. J Clin Endocrinol Metab common genetic variants associated
randomized, double-blind, placebo- 82:25782585 with serum DHEAS levels suggest a
controlled study. Neurology key role in ageing mechanisms.
Zhai G, Teumer A, Stolk L, Perry JR, PLoS Genet 7: e1002025
60:10711076 Vandenput L, Coviello AD, Koster A,
Yakin K, Urman B (2011) DHEA as a Bell JT, Bhasin S, Eriksson J, Zwain IH, Yen SS (1999)
miracle drug in the treatment of Eriksson A, Ernst F, Ferrucci L, Neurosteroidogenesis in astrocytes,
poor responders; hype or hope? Frayling TM, Glass D, Grundberg E, oligodendrocytes, and neurons of
Hum Reprod 26:19411944 Haring R, Hedman AK, Hofman A, cerebral cortex of rat brain.
Kiel DP, Kroemer HK, Liu Y, Endocrinology 140:38433852
Yen SS, Morales AJ, Khorram O (1995)
Replacement of DHEA in aging Lunetta KL, Maggio M, Lorentzon M,
men and women. Potential remedial Mangino M, Melzer D, Miljkovic I,

458
Chapter
The state-of-the-art in the development of

21 selective androgen receptor modulators


Ravi Jasuja, Mikhail N. Zacharov, and Shalender Bhasin

21.1 Introduction 459 21.6 Preclinical experience with first-generation


21.2 Structural classes of SARMs 460 non-steroidal SARMs 465
21.3 Structure-activity relationships of steroidal 21.7 Early phase I and II SARM trials 466
SARMs 460 21.8 Regulatory challenges to SARM
21.4 Non-steroidal SARMs 461 development 466
21.5 Mechanisms of tissue-selective actions 21.9 Key messages 467
of SARMs 463 21.10 References 467

21.1 Introduction other medical conditions such as anemia, osteoporosis,


Selective androgen receptor modulators (SARMs) are a autoimmune rheumatic disorders and androgen
class of androgen receptor (AR) ligands that bind the replacement in individuals at high risk of prostate
androgen receptor and display tissue-specific activation cancer, and for male contraception and to promote
of AR signaling (Bhasin et al. 2006; Narayanan et al. cutaneous wound healing (Bhasin et al. 2006; Narayanan
2008; Bhasin and Jasuja 2009). The impetus for the et al. 2008; Bhasin and Jasuja 2009).
development of SARMs was instigated by observations Structurally, SARMs can be categorized into ster-
that testosterone administration has potential beneficial oidal and non-steroidal SARMs. Many potent steroidal
effects on the skeletal muscle, bone, erythropoiesis, SARMs were developed during the 1940s and 1950s by
mood and sexual function, and that the anabolic effects modifying the chemical structure of the testosterone
of testosterone are dose related. The therapeutic appli- molecule. The pioneering efforts of scientists at Ligand
cations are limited by uncertainty about the potential Pharmaceuticals and the University of Tennessee
adverse effects of testosterone on cardiovascular events ushered in the modern era of non-steroidal SARMs
and the prostate (Bhasin et al. 2006; Narayanan et al. (Bhasin et al. 2006; Narayanan et al. 2008; Bhasin and
2008; Bhasin and Jasuja 2009). Therefore, pharmaco- Jasuja 2009). The Ligand team was the first to develop
phores that can activate androgen signaling and achieve a series of cyclic quinolinones that had anabolic activity
beneficial therapeutic effects without the potential on the skeletal muscle, and tissue selectivity (Edwards
adverse effects would be attractive as therapeutic agents et al. 1998; Hamann et al. 1999; van Oeveren et al.
for a variety of clinical disorders. Although much of the 2006; 2007; Miner et al. 2007; Higuchi et al. 2007).
SARM discovery effort is currently focused on their Dalton and Miller, at the University of Tennessee,
development as function-promoting anabolic therapies discovered that aryl-propionamides with structural
for limitations associated with sarcopenia (age-related similarities to bicalutamide and hydroxyflutamide
loss of muscle mass and strength), frailty, acute illness could activate AR-dependent transcriptional activity
and other chronic conditions, such as COPD, end-stage and had preferential anabolic effects on the muscle
renal disease, AIDS wasting syndrome and many types relative to the prostate (Dalton et al. 1998; He et al.
of cancers, they may also be useful in the treatment of 2002). Since then, most of the major pharmaceutical

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

459
Chapter 21: Development of SARMs

Structure-activity Compounds Chemical structure Fig. 21.1 Structure-activity relationship


relationship of steroidal SARMs. Reproduced with
permission from Bhasin and Jasuja (2009).
Removing 19-methyl 19-nortestosterone OH
increases anabolic (nandrolone) series
activity of compounds

O
19-nortestosterone

OH
17- alkyl Many orally active CH3 CH3
substitutions retard steroidal androgens
first-pass have 17- alkyl CH3 H
presystemic substitutions H H
metabolism
O

17-methyltestosterone
17- alkyl 7-methyl, CH3
OH

substitutions 9-nortestosterone
increase anabolic
activity

R
7-alkyl,19-nortestosterone

Esterification of Testosterone OCO(CH2)5CH3


CH3 H
17-hydroxyl group enanthate, cypionate
CH3
increases and undecanoate H

hydrophobicity and H H
extends duration of O
C26H40O3 MW 400.6
in vivo action
Testosterone enanthate

houses and many biotechnology companies have sup- and partial agonists. Most of the SARMs that have
ported SARM discovery programs. The development of advanced to phase I and II human trials are partial
non-steroidal SARMs during the past 15 years has been agonists: they are potent agonists in the muscle and
guided by a discovery approach that has been based on bone, but weak agonists in the prostate (Bhasin et al.
high-throughput screening of pharmacophore libraries 2006; Narayanan et al. 2008; Bhasin and Jasuja 2009).
using assays that are steered by pharmacophore binding
to AR and transactivation of AR-dependent reporter
genes. This approach has differed substantially from 21.3 Structure-activity relationships
that used in the discovery of the steroidal SARMs in of steroidal SARMs
the 1940s and 1950s, which was based on an under- While the structure-activity relationships of the
standing of the structure-activity relationships. As steroidal SARMs were elucidated seven decades ago
pharmaceutical and biotechnology companies have (Bhasin et al. 2006; Bhasin and Jasuja 2009; Hoffman
led the developmental effort related to the discovery of et al. 2009), only limited information about non-
non-steroidal SARMs, much of this research remains steroidal SARMs has been published (Fig. 21.1).
unpublished and unavailable for critical peer appraisal. 17a-alkyl substitution by retarding the presystemic
metabolism of testosterone extends its half-life and
makes it orally active (Dimick et al. 1961; Bhasin and
21.2 Structural classes of SARMs Jasuja 2009; Hoffman et al. 2009). Thus, a number of
Structurally, SARMs can be classified into steroidal oral androgens, such as 17a-methyltestosterone (Fig.
and non-steroidal. Functionally, these compounds 21.1), have 17a-alkyl substitution (Dimick et al. 1961).
can be categorized as pure agonists, pure antagonists However, when administered orally, 17a-alkylated

460
Chapter 21: Development of SARMs

androgens are potentially hepatotoxic and markedly Pharmaceuticals), tetrahydroquinoline analogs (Kaken
lower plasma HDL-C (Bhasin and Jasuja 2009; Pharmaceuticals, Inc.), benzimidazole, imidazolopyr-
Hoffman et al. 2009). azole, indole and pyrazoline derivatives (Johnson and
The nandrolone series of molecules was synthesized Johnson), azasteroidal derivatives (Merck), and anil-
based on the insight that the removal of the 19-methyl ine, diaryl aniline and benzoxazepinones derivatives
group increases the anabolic activity of testosterone (GSK) (Fig.21.2; Bhasin et al. 2006; Narayanan et al.
(Murad and Haynes 1980; Fragkaki et al. 2009). Thus, 2008; Bhasin and Jasuja 2009). This list is not exhaust-
a number of anabolic steroids are based on the 19-nor- ive; additional structural categories surely exist but
testosterone backbone. Nandrolone is reduced by the have not been published. We refer those who are
steroid 5a-reductase enzyme in target tissues to a less interested in SARM structures to an excellent treatise
potent androgen, dihydronandrolone. However, it on this topic by Narayanan et al. (2008).
serves only as a weak substrate for the CYP19 aromatase Ligand compounds LGD-2226 and LGD-2941
enzyme and is less susceptible to aromatization. are bicyclic 6-anilino quinolinone derivatives (van
7a-alkyl substitutions make testosterone less Oeveren et al. 2006; 2007; Miner et al. 2007). In
susceptible to 5a-reduction and increase its tissue select- the Hershberger assay using the castrated male rat
ivity with respect to the prostate (Bhasin and Jasuja (Hartsook and Hershberger 1961), these compounds
2009). Thus, 7a-methyl, 19-nortestosterone has anabolic stimulate the growth of levator ani muscle while
activity in the levator ani assay, but has a lower level exerting little effect on prostate size (Gao et al.
of prostate effects (Kumar et al. 1992; Sundaram et al. 2004a; 2004b; 2005; Miner et al. 2007). These com-
1994). A number of potent anabolic molecules in this pounds also increase bone mass and strength, and
series with varying alkyl groups have been investigated. bone architecture in ovariectomized female rats
Testosterone is cleared rapidly from circulation (Rosen and Negro-Vilar 2002; Kearbey et al. 2007;
and has a short half-life. The esterification of the Miner et al. 2007). LGD-2226 maintains male repro-
17b-hydroxyl group makes the molecule more hydro- ductive behavior in the castrated rat model (Miner
phobic and thereby retards its absorption from an oily et al. 2007).
depot injection. The longer the ester, the greater is The team led by Dalton and Miller at the University
its hydrophobicity, and the slower is the absorption of Tennessee developed novel SARMs, S1 and S4,
of the ester from its oily depot into the circulation. by structural modifications of aryl-propionamide
When 17b-hydroxyl esters of testosterone are injected analogs bicalutamide and hydroxyflutamide. These
intramuscularly in an oily suspension, they are relea- compounds S1 and S4 bind AR with high affinity
sed slowly from the oil depot into the circulation and demonstrate tissue selectivity in the Hershberger
(Snyder and Lawrence 1980; Sokol et al. 1982). The assay (Yin et al. 2003a; 2003b). When given at
slow release of 17b-hydroxyl esters from the oil depot an appropriate dose, S4 fully restores the levator ani
extends their duration of action. However, de- weight, muscle strength, BMD, bone strength and
esterification of testosterone esters is not rate- lean body mass, and suppresses LH and FSH (Gao
limiting; the half-life of testosterone enanthate in et al. 2004a; 2004b; 2005; Kearbey et al. 2007), while
plasma is similar to that of non-esterified testoster- only partially restoring prostate weight. S4 also pre-
one. Similarly, esterification of nandrolone to form vents ovariectomy-induced bone loss in a female rat
nandrolone decanoate increases its half-life. model of osteoporosis (Kearbey et al. 2007).
Oxandrolone is an oral androgen derived from DHT The hydantoin derivatives, developed by the BMS
that has a 17a-methyl substituent. The substitution of a group (Hamann et al. 2007) have an A-ring structure
second carbon with oxygen increases the stability of the that is similar to that of bicalutamide. The cyano or
3-keto group and increases its anabolic activity. It does nitro group of these molecules interacts with Q711
not undergo aromatization to an estrogen. and R752 (Manfredi et al. 2007; Ostrowski et al.
2007). The benzene ring or the naphthyl group,
together with the hydantoin ring, overlaps the steroid
21.4 Non-steroidal SARMs plane, while the hydantoin ring nitrogen forms an
Many structural categories of SARM pharmacophores H bond with N705. BMS-564929 binds AR with high
have been developed: aryl-propionamide (GTX, Inc.), affinity and high specificity (Manfredi et al. 2007;
bicyclic hydantoin (BMS), quinolinones (Ligand Ostrowski et al. 2007). BMS-564929 demonstrated

461
Chapter 21: Development of SARMs

Chemotype Structure Examples


Aryl-propionamide R Ostarine, andarine
X O
analogs
Y N Z
H OH

Bicyclic hydantoin HO O BMS-564929


analogs H

N R3
N

O R1 R2

Quinolinones R1 R2 LGD-2226, LGD-2941

O N N R4
H H R3

Tetrahydroquinoline R3 Kanen Pharmaceuticals,


analogs X S-40503
R1

Z
R2 N Y R4
H

Benzimidazoles Johnson and Johnsons


Cl N R benzimadozole
derivative
CF3
Cl N OH
H

Butanamides F O Merck SARM based on


butanamide scaffold
N
H

CF3

Fig. 21.2 Various structural classes of non-steroidal SARMs. Reproduced with permission from Bhasin and Jasuja (2009).

anabolic activity in the levator ani muscle, and a displayed high AR affinity and strong agonist activity
high degree of tissue selectivity as indicated by a in the prostate and levator ani, they demonstrated
substantially higher ED50 for the prostate than for little selectivity between androgenic and anabolic
levator ani muscle (Hamann et al. 2007; Manfredi tissues (Hanada et al. 2003). Significant in-vivo
et al. 2007; Ostrowski et al. 2007). Hydantoin deriva- pharmacological activity was only observed at high
tives are potent suppressors of LH. BMS-564929 is subcutaneous doses (Hanada et al. 2003).
orally available in humans, with a half-life of 814 Merck scientists have developed a number of
hours (Manfredi et al. 2007; Ostrowski et al. 2007). 4-azasteroidal derivatives and butanamides with ana-
Hanada et al. (2003) at Kaken Pharmaceutical Co. bolic activity in the muscle and bone and reduced
reported a series of tetrahydroquinoline derivatives as activities in reproductive tissues and sebaceous glands
AR agonists for bone. Although these compounds (Schmidt et al. 2009). One such SARM, TFM-4AS-1,

462
Chapter 21: Development of SARMs

induces the expression of some androgen-dependent antagonists and SARMs are reflected in conforma-
genes similar to DHT, and others to a lesser extent tionally distinct states with distinct thermodynamic
than DHT or not at all (Schmidt et al. 2009). TFM- partitioning. Differences in ligand-specific receptor
4AS-1 does not promote the conformation required conformation and proteinprotein interactions could
for interaction between the N-terminal and C-ter- result in tissue-specific gene regulation, due to poten-
minal domain, and displays tissue-selective agonistic tial changes in interactions with androgen response
activity on the muscle and bone while exhibiting elements (AREs), co-regulators or transcription
reduced activity in the prostate and sebaceous glands factors. Ligand-induced proteinprotein interactions
(Schmidt et al. 2009). The scientists at Johnson and contribute to interactions between the amino and
Johnson replaced the propionamide linker with cyclic carboxyl terminal ends of the AR (i.e. N/C inter-
elements such as pyrazoles, benzimidazoles, indoles action) and coactivator recruitment (Masiello et al.
and cyclic propionanilide mimetics (Ng et al. 2007). 2004). Both interactions are mediated by the inter-
Additional compounds have been developed by other action between the AF2 region of AR and the FxxLF
pharmaceutical companies (Narayanan et al. 2008). or LxxLL binding motifs (Song et al. 2003). The
hydrophobic groove present in the AF2 region of
21.5 Mechanisms of tissue-selective AR ligand-binding domain (AR-LBD) appears to be
more favorable for phenylalanine binding, which sug-
actions of SARMs gests that the N/C interaction is preferred and may
The expression profiles of androgen-dependent genes even be essential for agonist activity (Song et al. 2003;
induced by tissue-selective SARMs differ from those Narayanan et al. 2008).
induced by DHT (Narayanan et al. 2008; Schmidt Jasuja et al. (2009) used a variety of biophysical
et al. 2009). For instance, Narayanan et al. (2008) techniques, including steady-state second-derivative
found that an aryl-propionamide SARM, S-22, and absorption and emission spectroscopy, pressure and
DHT differed significantly from each other in the temperature perturbations, and 4,40 -bis-anilino-
recruitment of AR and its co-regulators to PSA naphthalene 8-sulfonate (bis-ANS) partitioning to
enhancer. S-22 also differed from DHT in that it determine the kinetics and thermodynamics of
induced rapid phosphorylation of several kinases the conformational changes in AR-LBD after DHT
(Narayanan et al. 2008). Three general hypotheses binding. The results of these biophysical experiments
have been proposed to explain the mechanisms that are consistent with the conclusion that DHT binding
contribute to tissue-specific transcriptional activation leads to energetic stabilization of AR-LBD, and sub-
and selectivity of the biological effects of SARMs stantial rearrangement of residues distant from the
(Bhasin et al. 2006; Narayanan et al. 2008; Bhasin ligand-binding pocket of AR (Jasuja et al. 2009).
and Jasuja 2009). These hypotheses are not mutually Dihydrotestosterone binding to AR-LBD involves
exclusive and it is possible that all three may be biphasic receptor rearrangement including the popu-
operative. lation of a molten globule-like intermediate state
The coactivator hypothesis assumes that the rep- (Fig.21.3; Jasuja et al. 2009). Although non-steroidal
ertoire of co-regulator proteins that associates with SARM-bound AR-LBD conformation has not been
the SARM-bound AR differs from that associated well characterized, Sathya et al. (2003) reported that
with testosterone-bound AR, leading to transcrip- some steroidal SARMs that have agonist activity in
tional activation of a differentially regulated set of vitro induce an activating conformational change
genes. According to the conformational hypothesis, without facilitating N/C interactions. These data sug-
ligand binding induces specific conformational gest that ligand-specific conformational change is
changes in the ligand-binding domain (LBD), which achievable with synthetic ligands.
modulates the surface topology and subsequent pro- Bohl et al. (2007) reported that bicalutamide
teinprotein interactions between AR and other co- adopts a greatly bent conformation in the AR.
regulators involved in genomic transcriptional acti- Although the A-ring and amide bond of the bicaluta-
vation or cytosolic proteins involved in non-genomic mide molecule overlaps with the steroidal plane, the
signaling (Bhasin et al. 2006; Narayanan et al. 2008; B-ring of bicalutamide folds away from the plane,
Bhasin and Jasuja 2009). Accordingly, the functional pointing to the top of the ligand binding pocket
differences among the three ligand classes agonists, (LBP), which forms a unique structural feature of this

463
Chapter 21: Development of SARMs

(A) Ligand-bound (C) Residue mobility difference 1.0 A

1.0 A

(B) Apo-form (D) Residue group dynamics

15
Non-CoaBC

CoaBC
10
RMSD (A)

CoaRS2

5 CoaRS1

LBI

0
0 2 4 6 8 10
Time (ns)

Fig. 21.3 See plate section for color version. Molecular modeling of AR-LBD upon ligand binding. The simulations were performed at
295.5 K and atmospheric pressure. (A, B) Superimposed representations of AR-LBD in the ligand-bound form and apo-form, respectively.
Pink coloring shows the initial structures, and the pale green coloring shows the structures after 10 ns of simulations. (C) The AR-LBD apo-form
colored by the difference between standard deviations of residue RMSDs (root mean square deviations) for LBD-R1881 complex and
apo-form over 610 ns. Positive values correspond to greater mobility in the apo-form. (D) The predicted RMSDs for ligand-binding interface
(LBI), coactivator recruitment surface 1 (CoaRS1), coactivator recruitment surface 2 (CoaRS2) and coactivator binding cleft (CoaBC). Red curves
show RMSDs of helices in a ligand-bound structure, green curves in an apo-form structure. The groups of residues are defined as follows:
LBI (V685, L701, N705, L707, Q711, L744, M745, M749, R752, Y763, F764, Q783, M787, F876, T877, L880, F891, M895), CoaRS1 (V713, V730, M734),
CoaRS2 (E709, L712, V713, V715, V716, K717, K720, F725, R726, V730, Q733, M734, I737, Q738), CoaBC (helices 3, 30 , 4, 5, 12), Non-CoaBC
(helices 1, 6, 7, 8, 9, 10, 11). Reproduced with permission from Jasuja et al. (2009).

class of ligands (Bohl et al. 2007). The A-ring cyano N705, mimicking ring C and the 17b-OH group in
group forms H bonds with Q711 and R752, similar to 5a-DHT (Bohl et al. 2007); these H bonding inter-
the 3-keto group in 5a-DHT (Bohl et al. 2007). The actions are critical for high binding affinity. Favorable
chiral hydroxyl group forms H bonds with L704 and hydrogen bonding between ligand and the T877 side

464
Chapter 21: Development of SARMs

chain, structural features that mimic the 3-keto group either the steroid 5a-reductases or the CYP19 aroma-
of testosterone, and hydrophobic interactions are crit- tase. Some differences of the actions of SARMs from
ical for the ligand to bind with high affinity and those of testosterone could be related to the inability
stimulate AR action (Bohl et al. 2007). The elucida- of non-steroidal SARMs to undergo 5a-reduction or
tion of the X-ray crystal structure of S1-bound AR has aromatization.
shown that W741 side chain is displaced by the B-ring The tissue selectivity of SARMs could also be
to expand the binding pocket so that the compound related to differences in their tissue distribution. Auto-
orients towards the AF2 region (Bohl et al. 2007). radiography studies with bicalutamide and hydantoin
The protein fold of the SARM-bound AR is the same derivatives have not revealed the preferential accumu-
for steroidal and non-steroidal SARMs (Bohl et al. lation of these SARMs in anabolic tissues (Hamann
2007). It remains unclear how ligandreceptor inter- 2004). Differences in pharmacokinetics and metabolic
action determines the agonist or antagonist activity clearance might also contribute to disparities in the in-
of the ligand. vivo activities of SARMs with similar in-vitro binding
The unique conformation imparted by specific affinity and transactivational potency.
ligand-binding events allows the recruitment of a
specific repertoire of AR co-regulator proteins which 21.6 Preclinical experience with
contribute to tissue-specificity of action. Tissue select-
ivity of SARMs might also be related to tissue- first-generation non-steroidal SARMs
specific expression of co-regulator proteins (Heinlein Preclinical proof-of-concept studies with a number of
and Chang 2002). We have investigated the molecular non-steroidal SARMs have revealed promising tissue
events that follow coactivator binding to AR, and the selectivity (Gao et al. 2004a; 2004b; Bhasin et al. 2006;
mechanisms that govern the sequence-specific effects Kearbey et al. 2007; Miner et al. 2007; Narayanan et al.
of AR co-regulators are poorly understood (Zacharov 2008; Bhasin and Jasuja 2009). Much of the preclinical
et al. 2011). Using consensus coactivator sequence data generated by pharmaceutical companies has
D11-FxxLF and biophysical techniques, we showed remained unpublished; therefore, it is difficult to com-
that coactivator association is followed by conforma- pare the relative potency and tissue selectivity of dif-
tional rearrangement in AR-LBD that is enthalpically ferent SARMs, and the examples cited below from the
and entropically favorable, with activation energy of published literature should not be viewed as evidence
29.8  4.2 kJ/mol (Zacharov et al. 2011). Each coac- of exclusive or unique activity of a particular SARM.
tivator induces a distinct conformational state in However, it is apparent that many non-steroidal
the dihydrotestosterone:AR-LBD:coactivator complex SARMs have achieved varying degrees of selectivity of
(Zacharov et al. 2011). Computational modeling anabolic action on the muscle relative to prostate
revealed that the intramolecular rearrangements in and seminal vesicles (Gao et al. 2004a; 2004b; Bhasin
AR-LBD backbone induced by the specific coactiva- et al. 2006; Kearbey et al. 2007; Miner et al. 2007;
tors are different (Zacharov et al. 2011). These data Ostrowski et al. 2007; Narayanan et al. 2008; Bhasin
suggest that coactivators may impart specificity in the and Jasuja 2009; Schmidt et al. 2009). Although
transcriptional machinery by changing the steady- Hershberger assay based on the mass of levator ani
state conformation of AR-LBD. The data provide muscle in castrated rodents has been used typically to
direct evidence that, even in the presence of the same determine the relative anabolic activity of SARMs
ligand, AR-LBD can occupy distinct conformational (Hartsook and Hershberger 1961), some SARMs,
states depending on its interactions with specific coac- such as the Ligand compounds, LGD-2226, LGD-
tivators in the tissues; this coactivator-specific con- 2941 and LGD-4033, have been shown to stimulate
formational gating may dictate subsequent binding the growth of other skeletal muscle groups, such as
partners and interaction with the DNA-response the plantaris, gastrocnemius and biceps brachii, as
elements (Zacharov et al. 2011). well (Miner et al. 2007). LGD-2226, LGD-2941 and
Testosterone actions in some androgenic tissues LGD-4033 have also been shown to maintain measures
are amplified by its conversion to 5a-DHT (Russell of mating behavior in castrated male rats, including the
and Wilson 1994); while its effects on some other number of mounts, intromissions and ejaculations,
tissues require its aromatization to estradiol. The and copulatory efficiency (Miner et al. 2007). Several
non-steroidal SARMs do not serve as substrates for SARMs Ligand Pharmaceuticals LGD-2226,

465
Chapter 21: Development of SARMs

LGD-2941 and LGD-4033 compounds, GTXs reductions in HDL cholesterol. The suppression of
compound, ostarine, and Mercks compound, TFM- HDL-C might reflect the combined effects of the oral
4AS-1 have been reported to increase BMD, bone route of administration and the lack of aromatization.
mass and strength, and restore bone architecture in It is possible that a systemic route of administration
ovariectomized female rodents (Miner et al. 2007; transdermal or intramuscular might attenuate the
Kearbey et al. 2007; Schmidt et al. 2009). In intact potential for HDL-C reductions.
rats, SARMs with agonist activity would be expected Most of the first-generation non-steroidal SARMs
to inhibit LH and FSH and spermatogenesis, as has are partial agonists (Gao et al. 2004a; 2004b): they
been reported for the S4 compound. compete with endogenous testosterone and, in intact
animals, can antagonize the effects of testosterone on
the prostate. Such SARMs with antagonistic or low
21.7 Early phase I and II SARM trials intrinsic activity in prostate might be useful in the
A number of first-generation SARMs have advanced treatment of BPH or prostate cancer. The suppressive
to phase I trials, and some are in phase II trials effects of this class of SARMs on gonadotropin secre-
(Basaria et al. 2012). At the doses that have been tion could have potential application for the develop-
tested, the first-generation SARMs have been reported ment of male contraceptives (Gao et al. 2004a;2004b).
to induce modest gains of 1.0 to 1.5 kg in fat-free Because the currently available SARMs are neither
mass and muscle strength, and small reductions in fat aromatized nor 5a-reduced, these compounds would
mass. At these doses, these compounds on which data face an uphill regulatory bar for approval as they
have been reported in public forums have generally would be required to show efficacy and safety in many
been well tolerated with minimal or no changes in more domains of androgen action than has been
liver enzymes and blood counts, and modest reduc- required of testosterone formulations.
tions in HDL cholesterol and triglycerides. However, The ability of SARMs to promote both muscle
it is possible that the next generation of SARM mol- strength and bone mechanical strength constitutes a
ecules will have greater potency and selectivity than unique advantage over other therapies for osteopor-
the first-generation SARMs. osis that only increase bone density. By increasing
These compounds are being positioned for early muscle mass and strength, SARMs might also reduce
efficacy trials for cancer cachexia, aging-associated func- fall propensity and thereby reduce fracture risk. Fur-
tional limitations, sarcopenia, osteoporosis and frailty. thermore, the regulatory pathway for the approval of
Also, SARMs that potently inhibit gonadotropins, drugs for osteoporosis has been well delineated
but spare the prostate, would be attractive as candi- because of precedents set by previously approved
dates for male contraception. The use of SARMs for drugs. In contrast, the pathway for approval of func-
the treatment of androgen-deficiency syndromes in tion-promoting anabolic therapies has not been
men has been proposed; the relative advantages of clearly established.
SARMs over testosterone for this indication are not Considerable effort is underway to generate a
readily apparent. Many biological functions of testos- consensus around indications, efficacy outcomes in
terone, especially its effects on libido and behavior, pivotal trials, and minimal clinically important differ-
bone, and plasma lipids require its aromatization to ences in key efficacy outcomes; these efforts should
estrogen; because the currently available SARMs are facilitate efficacy trials of candidate molecules. In view
neither aromatized nor 5a-reduced, these compounds of these uncertainties, the first-generation SARMs are
would face an uphill regulatory bar for approval as being positioned for acute or subacute indications
they would be required to show efficacy and safety in for grievous conditions, such as rehabilitation after a
many more domains of androgen action than has hip fracture or an acute illness, and cancer cachexia.
been required of testosterone formulations. Efficacy trials with short intervention durations for
these acute and subacute indications would provide a
21.8 Regulatory challenges to SARM substantially more favorable risk-to-benefit ratio than
long-term trials for the prevention of more chronic
development conditions such as sarcopenia. The initial approval for
In phase I and II trials, oral administration of the short-term indications would also allow accumulation
first-generation SARMs has been associated with of safety data in clinical populations.

466
Chapter 21: Development of SARMs

21.9 Key messages


repertoire of co-regulator proteins to the liganded
 Non-steroidal SARMs are being developed as a receptor, and thereby induce ligand-specific gene
new class of function-promoting anabolic expression. The inability of non-steroidal SARMs to
therapies for functional limitations associated undergo 5a-reduction and aromatization, and
with frailty, acute and chronic illnesses, aging, differences in the tissue-specific expression of
cancer and cachexia, anemia and osteoporosis. co-regulator proteins, may also contribute to their
 The first-generation non-steroidal SARMs are tissue selectivity.
partial agonists and have tissue-selective  The efficacy trials of SARMs are just beginning,
anabolic effects on the muscle and bone versus and initial trials have shown early evidence of
the reproductive tissues. safety and modest gains in lean body mass.
 The mechanisms of tissue-selective actions of  Initial efficacy trials of SARMs are likely to be for
SARMs are incompletely understood. Non-steroidal short-term treatment indications for acute or
SARMs induce unique conformational changes in subacute conditions associated with functional
the AR ligand-binding domain, and recruit a unique limitations.

21.10 References Discovery of nonsteroidal


androgens. Biochem Biophys Res
Gao W, Kearbey JD, Nair VA,
Chung K, Parlow AF, Miller DD,
Basaria S, Collins L, Dillon EL, Commun 244:14 Dalton JT (2004b) Comparison of
Orwoll K, Storer TW, Miciek R, the pharmacological effects of a
Ulloor J, Zhang A, Eder R, Dimick DF, Heron M, Baulier EE,
Jaylee MF (1961) A comparative novel selective androgen receptor
Zientek H, Gordon G, Kazmi S, modulator, the 5alpha-reductase
Sheffield-Moore M, Bhasin S (2012) study of the metabolic fate of
testosterone, 17 alpha-methyl- inhibitor finasteride, and the
The safety, pharmacokinetics, and antiandrogen hydroxyflutamide in
the effects of LGD-4033, a novel testosterone. 19-nor-testosterone.
17 alpha-methyl-19-nor- intact rats: new approach for benign
non-steroidal oral, selective prostate hyperplasia. Endocrinology
androgen receptor modulator testosterone and 17 alpha-
methylestr-5(10)-ene-17 beta-ol-3- 145:54205428
(SARM), in healthy young men.
J Gerontol A Biol Sci Med Sci; in press one in normal males. Clin Chim Gao W, Reiser PJ, Coss CC,
Acta 6:6371 Phelps MA, Kearbey JD, Miller DD,
Bhasin S, Jasuja R (2009) Selective Dalton JT (2005) Selective androgen
Edwards JP, West SJ, Pooley CL,
androgen receptor modulators as receptor modulator treatment
Marschke KB, Farmer LJ, Jones TK
function promoting therapies. improves muscle strength and
(1998) New nonsteroidal androgen
Curr Opin Clin Nutr Metab body composition and
receptor modulators based on
Care 12:232240 prevents bone loss in
4-(trifluoromethyl)-2(1H)-
Bhasin S, Calof OM, Storer TW, Lee ML, pyrrolidino[3,2-g] quinolinone. orchidectomized rats.
Mazer NA, Jasuja R, Montori VM, Bioorg Medl Chem Lett 8:745750 Endocrinology 146:48874897
Gao W, Dalton JT (2006) Drug Hamann LG (2004) Discovery and
Fragkaki AG, Angelis YS,
insight: testosterone and selective preclinical profile of a highly potent
Koupparis M, Tsantili-Kakoulidou
androgen receptor modulators as and muscle selective androgen
A, Kokotos G, Georgakopoulos C
anabolic therapies for chronic illness receptor modulator (SARM).
(2009) Structural characteristics of
and aging. Nat Clin Pract Endocrinol 227th National Meeting of the
anabolic androgenic steroids
Metab 2:146159 American Chemical Society
contributing to binding to the
Bohl CE, Wu Z, Miller DD, Bell CE, androgen receptor and to their Medicinal Chemistry Division,
Dalton JT (2007) Crystal structure anabolic and androgenic activities. March 28April 1, Anaheim, CA
of the T877A human androgen Applied modifications in the steroidal Hamann LG, Mani NS, Davis RL,
receptor ligand-binding domain structure. Steroids 74:172197 Wang XN, Marschke KB, Jones TK
complexed to cyproterone acetate Gao W, Reiser PJ, Kearbey JD, Phelps (1999) Discovery of a potent, orally
provides insight for ligand-induced MA, Coss CC, Miller DD, Dalton JT active, nonsteroidal androgen
conformational changes and (2004a) Effects of Novel Selective receptor agonist: 4-ethyl-1,2,3,4-
structure-based drug design. J Biol Androgen Receptor Modulator tetrahydro-6-(trifluoromethyl)-8-
Chem 282:1364813655 (SARM) on Skeletal Muscle Mass and pyridono[5,6-g]-quinoline
Dalton JT, Mukherjee A, Zhu Z, Strength in Castrated Male Rats. The (LG121071). J Med Chem
Kirkovsky L, Miller DD (1998) Endocrine Society, New Orleans 42:210212

467
Chapter 21: Development of SARMs

Hamann LG, Manfredi MC, Sun C, human growth hormone use. function with reduced impact on
Krystek SR Jr, Huang Y, Bi Y, J Strength Cond Res 23(5 Suppl): prostate. Endocrinology 148:363373
Augeri DJ, Wang T, Zou Y, S1S59 Murad F, Haynes RC (1980)
Betebenner DA, Fura A, Seethala R, Jasuja S, Ulloor J, Yengo CM, Androgens and anabolic steroids.
Golla R, Kuhns JE, Lupisella JA, Choong K, Istomin AY, Livesay DR, In: Goodman L, Gilman A (eds) The
Darienzo CJ, Custer LL, Price JL, Jacobs DJ, Swerdloff RS, Pharmacological Basis of
Johnson JM, Biller SA, Zahler R, Mikovsk J, Larsen RW, Bhasin S Therapeutics, 6th edn. McMilan
Ostrowski J (2007) Tandem (2009) Kinetic and thermodynamic Press, New York, pp 14481465
optimization of target activity and characterization of DHT-induced
elimination of mutagenic potential Narayanan R, Mohler ML, Bohl CE,
conformational perturbations in Miller DD, Dalton JT (2008)
in a potent series of N-aryl bicyclic androgen receptor ligand binding
hydantoin-based selective androgen Selective androgen receptor
domain. Mol Endocrinol modulators in preclinical and
receptor modulators. Bioorg Medl 23:12311241
Chem Lett 17:18601864 clinical development. Nucl Recept
Kearbey JD, Gao W, Narayanan R, Signal 6:e010
Hanada K, Furuya K, Yamamoto N, Fisher SJ, Wu D, Miller DD, Ng RA, Lanter JC, Alford VC,
Nejishima H, Ichikawa K, Dalton JT (2007) Selective androgen Allan GF, Sbriscia T, Lundeen SG,
Nakamura T, Miyakawa M, receptor modulator (SARM) Sui Z (2007) Synthesis of potent and
Amano S, Sumita Y, Oguro N treatment prevents bone loss tissue-selective androgen receptor
(2003) Bone anabolic effects of and reduces body fat in modulators (SARMs): 2-(2,2,2)-
S-40503, a novel nonsteroidal ovariectomized rats. Pharm trifluoroethyl-benzimidazole
selective androgen receptor Res 24:328335 scaffold. Bioorg Medl Chem Lett
modulator (SARM), in rat models
Kumar N, Didolkar AK, Monder C, 17:17841787
of osteoporosis. Biol Pharm Bull
26:15631569 Bardin CW, Sundaram K (1992) Ostrowski J, Kuhns JE, Lupisella JA,
The biological activity of 7 Manfredi MC, Beehler BC, Krystek
Hartsook EW, Hershberger TV (1961) alpha-methyl-19-nortestosteroneis SR Jr, Bi Y, Sun C, Seethala R,
Effect of synthetic steroid or not amplified in male reproductive Golla R, Sleph PG, Fura A, An Y,
phenanthrene compounds upon tract as is that of testosterone. Kish KF, Sack JS, Mookhtiar KA,
weight gain and feed efficiency of Endocrinology 130:36773683 Grover GJ, Hamann LG (2007)
rats. ProcSocExp Biol Med 106:5760
Manfredi MC, Bi Y, Nirschl AA, Pharmacological and X-ray
He Y, Yin D, Perera M, Kirkovsky L, Sutton JC, Seethala R, Golla R, structural characterization of a
Stourman N, Li W, Dalton JT, Miller Beehler BC, Sleph PG, Grover GJ, novel selective androgen receptor
DD (2002) Novel nonsteroidal Ostrowski J, Hamann LG (2007) modulator: potent hyperanabolic
ligands with high binding affinity Synthesis and SAR of stimulation of skeletal muscle with
and potent functional activity for the tetrahydropyrrolo[1,2-b][1,2,5] hypostimulation of prostate in rats.
androgen receptor. Eur J Med Chem thiadiazol-2(3H)-one 1,1-dioxide Endocrinology 148:412
37:619634 analogues as highly potent selective Rosen J, Negro-Vilar A (2002) Novel,
Heinlein CA, Chang C (2002) androgen receptor modulators. non-steroidal, selective androgen
Androgen receptor (AR) Bioorg Medl Chem Lett 17:44874490 receptor modulators (SARMs) with
coregulators: an overview. Endocr Masiello D, Chen SY, Xu Y, anabolic activity in bone and muscle
Rev 23:175200 Verhoeven MC, Choi E, Hollenberg and improved safety profile.
Higuchi RI, Arienti KL, Lopez FJ, Mani AN, Balk SP (2004) Recruitment of J Musculoskelet Neuronal Interact
NS, Mais DE, Caferro TR, Long YO, beta-catenin by wild-type or mutant 2:222224
Jones TK, Edwards JP, Zhi L, androgen receptors correlates with Russell DW, Wilson JD (1994) Steroid
Schrader WT, Negro-Vilar A, ligand-stimulated growth of 5 alpha-reductase: two genes/two
Marschke KB (2007) Novel series of prostate cancer cells. Mol Endocrinol enzymes. Annu Rev Biochem
potent, nonsteroidal, selective 18:23882401 63:2561
androgen receptor modulators Miner JN, Chang W, Chapman MS, Sathya G, Chang CY, Kazmin D,
based on 7H-[1,4]oxazino[3,2-g] Finn PD, Hong MH, Lopez FJ, Cook CE, McDonnell DP (2003)
quinolin-7-ones. J Med Chem Marschke KB, Rosen J, Schrader W, Pharmacological uncoupling of
50:24862496 Turner R, van Oeveren A, androgen receptor-mediated
Hoffman JR, Kraemer WJ, Bhasin S, Viveros H, Zhi L, Negro-Vilar A prostate cancer cell proliferation
Storer T, Ratamess NA, Haff GG, (2007) An orally active selective and prostate-specific antigen
Willoughby DS, Rogol AD (2009) androgen receptor modulator is secretion. Cancer Res
Position stand on androgen and efficacious on bone, muscle, and sex 63:80298036

468
Chapter 21: Development of SARMs

Schmidt A, Harada S-I, Kimmel DB, Beta-catenin binds to the activation studies on 6-bisalkylamino-2-
Bai C, Chen F, Rutledge SJ, function 2 region of the androgen quinolinones. Bioorg Med Chem
Vogel RL, Sccafonas A, Gentile MA, receptor and modulates the effects Lett 17:15271531
Nantermet PV, McElwee-Witmer S, of the N-terminal domain and TIF2
Yin D, Gao W, Kearbey JD, Xu H,
Pennypacker B, Massarachia P, on ligand-dependent transcription.
Chung K, He Y, Marhefka CA,
Sahoo SP, Kim Y, Meissner RS, Mol Cell Biol 23:16741687
Veverka KA, Miller DD, Dalton JT
Hartman GD, Duggan ME, Sundaram K, Kumar N, Bardin CW (2003a) Pharmacodynamics of
Rodan GA, Towler DA, Ray WJ (1994) 7 alpha-methyl-19- selective androgen receptor
(2009) Identification of anabolic nortestosterone: an ideal androgen modulators. J Pharmacol Exp Ther
selective androgen receptor for replacement therapy. 304:13341340
modulators with reduced activities Recent Prog Horm Res
in the reproductive tissues and Yin D, He Y, Perera MA, Hong SS,
49:373376
sebaceous glands. J Biol Chem Marhefka C, Stourman N,
284:3636736376 van Oeveren A, Motamedi M, Kirkovsky L, Miller DD, Dalton JT
Mani NS, Marschke KB, Lopez FJ, (2003b) Key structural features of
Snyder PJ, Lawrence DA (1980) Schrader WT, Negro-Vilar A, Zhi L nonsteroidal ligands for binding
Treatment of male hypogonadism (2006) Discovery of 6-N,N-bis and activation of the androgen
withtestosterone enanthate. (2,2,2-trifluoroethyl)amino-4- receptor. Mol Pharmacol
J Clin Endocrinol Metab trifluoromethylquinolin-2(1H)-one 63:211223
51:13351339 as a novel selective androgen
receptor modulator. J Med Chem Zacharov MN, Pillai BK, Bhasin S,
Sokol RZ, Palacios A, Campfield LA,
49:61436146 Ulloor J, Istomin AY, Guo C,
Saul C, Swerdloff RS (1982)
Godzik A, Kumar R, Jasuja R
Comparison of the kinetics of van Oeveren A, Motamedi M, (2011)) Dynamics of
injectable testosterone in eugonadal Martinborough E, Zhao S, Shen Y, coregulator-induced
and hypogonadal men. Fertil Steril West S, Chang W, Kallel A, conformational perturbations in
37:425430 Marschke KB, Lopez FJ, Negro- androgen receptor ligand binding
Song LN, Herrell R, Byers S, Shah S, Vilar A, Zhi L (2007) Novel selective domain. Mol Cell Endocrinol
Wilson EM, Gelmann EP (2003) androgen receptor modulators: SAR 341:18

469
Chapter
The essential role of testosterone

22 in hormonal male contraception


Eberhard Nieschlag and Hermann M. Behre

22.1 General prospects 470 22.4.2 Testosterone plus gonadotropin-


22.1.1 Reasons for male contraception 470 releasing hormone antagonists 475
22.1.2 Existing methods 471 22.5 Testosterone combined with progestins 476
22.1.3 New approaches to male 22.5.1 Testosterone or 19-nortestosterone plus
contraception 471 depot medroxyprogesterone
22.2 Principle of hormonal male acetate 476
contraception 471 22.5.2 Testosterone plus levonorgestrel 484
22.3 Testosterone alone 472 22.5.3 Testosterone plus cyproterone
22.3.1 Testosterone enanthate 472 acetate 484
22.3.2 Testosterone buciclate 473 22.5.4 Testosterone plus dienogest 485
22.3.3 Testosterone undecanoate 473 22.5.5 Testosterone plus desogestrel or
22.3.3.1 Oral testosterone etonogestrel 485
undecanoate 473 22.5.6 Testosterone plus
22.3.3.2 Testosterone undecanoate in 19-norethisterone 486
tea seed oil 473 22.6 Side-effects of hormonal male
22.3.3.3 Testosterone undecanoate in contraception 486
castor oil 473 22.7 Acceptability of hormonal male
22.3.4 Testosterone pellets 474 contraception 487
22.3.5 19-Nortestosterone 474 22.8 Outlook 487
22.3.6 7a-Methyl-19-nortestosterone 474 22.9 Key messages 487
22.4 Testosterone plus gonadotropin-releasing 22.10 References 488
hormone analogs 474
22.4.1 Testosterone plus gonadotropin-
releasing hormone agonists 474

22.1 General prospects


distribution and utilization of female contraceptive
22.1.1 Reasons for male contraception methods might ameliorate this situation, the contribu-
Female contraception is very effective. Nevertheless, tion of a male contraceptive is well worth considering.
40% of the 208 million pregnancies occurring world- Men enjoy the pleasures of sex, but can do little to
wide in 2008 were unplanned, and half of the unin- contribute to the tasks of family planning a pharma-
tended pregnancies were terminated by abortion; an cological male contraceptive is perhaps long overdue. In
intervention that ended fatally for about 250 000 of addition, the risks of contraception would also be more
these women (Singh et al. 2010). Although improved fairly shared between women and men. Representative

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

470
Chapter 22: Role of testosterone in male contraception

surveys have shown that a pharmacological male con- introduction of new materials (e.g. polyurethane) for
traceptive would be acceptable to large segments of the condoms, the inherent disadvantages of these methods
population in industrialized nations, and would thus preventing sperm transport into the female tract per-
contribute to further stabilization of population dynam- sist, and must be replaced and/or supplemented by
ics. It might also help developing countries whose expo- pharmacological methods. Post-testicular approaches
nential population growth endangers economic, social to male contraception are still in the preclinical phase
and medical progress. Last but not least, male contra- (Cooper and Yeung 2010). By investigating the
ception can be considered an outstanding issue in the molecular physiology of sperm maturation, epididy-
political field of gender equality. mal function and fertilization, the aim is to identify
processes that might be blocked by specific pharmaco-
22.1.2 Existing methods logical agents with rapid onset of action. However, all
Traditional male methods of contraception such as peri- substances investigated so far have shown toxic side-
odic abstinence or coitus interruptus are associated with effects when interfering effectively with sperm func-
a relatively high rate of unwanted pregnancy and also tion. At the moment then, only hormonal methods
cause a disturbance in sexual activity. Condoms are the fulfill most of the requirements for a male contracep-
oldest barrier method available. However, when using tive and are currently under clinical development.
condoms conception rates are relatively high, with 12 All hormonal male contraceptives clinically tested
out of 100 couples conceiving during the first year of use to date are based on testosterone, either on testoster-
(Pearl index 12). Condom use has increased since the one alone or on a combination of testosterone with
beginning of the AIDS epidemic, but more for protec- other hormones, in particular with either gestagens or
tion from HIV infection and other sexually transmitted GnRH analogs. Because of the essential role of testos-
diseases than for contraceptive purposes. terone, it is appropriate to include an overview on
Vasectomy is a safe and surgically relatively simple current hormonal approaches to male contraception
method for male contraception. The rate of unwanted in this volume on testosterone.
pregnancy after vasectomy is less than 1%. The draw-
back to vasectomy is that it is not easily reversible. 22.2 Principle of hormonal male
Achieving fatherhood after vasectomy requires either
surgical reversal or sperm extraction from a testicular
contraception
biopsy and intracytoplasmatic sperm injection into The testes have an endocrine and an exocrine function:
the ovum. Only about 50% of these men will become the production of androgens and of male gametes. Sup-
fathers in the end. pression of gamete production or interference with
Given the disadvantages of these mechanical male gamete function without affecting the endocrine func-
methods, what then are the prerequisites for an ideal tion is the goal of endocrine approaches to male fertility
male contraceptive (Nieschlag et al. 2010)? It should be: regulation. However, since the two functions of the testes
are interdependent, it has remained impossible so far to
 as effective as comparable female methods
suppress spermatogenesis exclusively and reversibly
 acceptable to both partners without significantly affecting androgen synthesis.
 rapidly effective Follicle-stimulating hormone and LH/testosterone
 independent of sexual act are responsible for the maintenance of fully normal
 free of side-effects, especially without influence on spermatogenesis (see Chapters 2 and 6; also Weinbauer
masculinity, libido and potency and Nieschlag 1996). If only one of the two is elimin-
 without influence on progeny ated, spermatogenesis will be reduced, but only in
 reversible in regard to fertility quantitative terms; i.e. fewer but normal sperm will
 easily available and financially affordable. be produced and azoospermia will not be achieved.
This has been demonstrated in monkeys by the elimin-
22.1.3 New approaches to male ation of FSH by immunoneutralization, resulting in
reduced sperm numbers but not in complete azoosper-
contraception mia (Srinath et al. 1983), which at least until quite
Despite attempts to improve the existing methods, e.g. recently was considered to be required for an effective
vas occlusion instead of surgical dissection, or the male method. Therefore, even if new modalities for the

471
Chapter 22: Role of testosterone in male contraception

selective suppression of FSH or FSH action should This leads to the general principle of hormonal male
become available, it remains doubtful whether they contraception: namely the suppression of FSH and LH,
would lead to a method for male contraception resulting in depletion of intratesticular testosterone and
(Nieschlag 1986). cessation of spermatogenesis, while, at the same time,
Until such results become available, the concept of peripheral testosterone is substituted with an androgen
azoospermia remains valid as a prerequisite for effective preparation. This can be achieved by testosterone
hormonal male contraception. However, as it is very alone. However, since testosterone alone does not
difficult to achieve azoospermia uniformly in all volun- lead to azoospermia or severe oligozoospermia
teers participating in clinical trials for hormonal male (<1 million/ml) in all individuals tested, testosterone
contraception, and the pregnancy rates appear to be needs to be combined with other substances suppressing
acceptably low if sperm counts drop below 1 million/ pituitary gonadotropin secretion. As in female hormo-
ml, investigators active in the field reached a consensus nal contraceptives, gestagens as pituitary-suppressing
that azoospermia or at least oligozoospermia i.e. <1 agents are being tested in men in combination with
million/ml sperm should be the goal for an effective androgens. Gonadotropin-releasing hormone agonists,
hormonal method (Nieschlag et al. 2007). To achieve as well as antagonists, are also being explored as further
this goal, not only FSH must be suppressed, but also possible combinations with androgens.
intratesticular testosterone must be drastically reduced.
Since testosterone alone can maintain spermatogenesis 22.3 Testosterone alone
and much lower testosterone concentrations appear to
be necessary for maintenance of spermatogenesis than 22.3.1 Testosterone enanthate
previously considered, intratesticular testosterone must According to the principle outlined above, testoster-
be depleted to such an extent that peripheral serum one should be the first choice for hormonal male
concentrations drop into the hypogonadal range. In contraception since it not only suppresses pituitary
order to maintain androgenicity, including libido, LH and FSH secretion, but also replaces testosterone.
potency, male sex characteristics, psychotropic effects, Indeed, since the 1970s various investigations have
protein anabolism, bone structure and hematopoiesis, been undertaken to suppress spermatogenesis with
testosterone levels in the general circulation have to be testosterone alone (Reddy and Rao 1972; Patanelli
replaced, while the testes themselves are depleted of 1978). However, these were all surrogate studies,
testosterone. However, even testes of volunteers achiev- looking for sperm counts and not for pregnancies or
ing azoospermia show measurable testosterone concen- rather for the prevention of pregnancies. Not until
trations, although reduced to 2% of normal, and 1990 was an initial study testing this form of male
volunteers developing azoospermia have low intrates- contraception published by the WHO: the first study
ticular levels similar to those suppressing only to oligo- ever performed on the efficacy of hormonal male
zoospermia (see Chapter 6). Therefore, other factors contraception (WHO 1990). Volunteers in 10 centers
must be of additional importance. Interestingly, the on four continents participated and received 200 mg
macaque monkey suppressed to azoospermia shows testosterone enanthate intramuscularly per week.
hardly any decrease in intratesticular testosterone, and Those volunteers developing azoospermia within the
elimination of FSH action appears to be more important first six months continued to receive injections for
than intratesticular testosterone (Weinbauer et al. 2001; a further year. In this period (efficacy phase), couples
Narula et al. 2002). For some time it was thought that the refrained from using any further contraceptive
intratesticular conversion of testosterone to DHT is of methods. A total of 137 men reached the efficacy
importance in the maintenance of spermatogenesis and phase. During this period only one pregnancy
should be interfered with. However, the application of a occurred. This high rate of efficacy is well comparable
5a-reductase inhibitor did not additionally affect the to that of established female methods. This was a very
suppression of spermatogenesis by testosterone alone encouraging result. However, only about two-thirds of
(McLachlan et al. 2000). Recently, the number of CAG all participants developed azoospermia. The other vol-
repeats in exon 1 of the androgen receptor has been unteers showed strong suppression of spermatogen-
found to determine the suppressibility of spermatogen- esis, as evidenced by oligozoospermia (Waites 2003).
esis, provided FSH and LH are well suppressed (von In order to answer the question of whether men
Eckardstein et al. 2002; Nieschlag et al. 2011). developing oligozoospermia can be considered

472
Chapter 22: Role of testosterone in male contraception

infertile, a second worldwide multicenter study in the early phase of development. However, even
followed (WHO 1996). In this study azoospermia when high doses of 3  80 mg were taken daily for
again proved to be a most effective prerequisite for 12 weeks, only one of seven volunteers developed
contraception. If sperm concentrations, however, azoospermia (Nieschlag et al. 1978). Although this
failed to drop below 3  106/ml, resulting pregnancy result was disappointing, the study demonstrated that
rates were higher than when using condoms. When stable levels of testosterone in serum are important to
sperm concentrations decreased below 3  106/ml, suppress pituitary gonadotropins.
which was the case in 98% of the participants, protec-
tion was not as effective as for azoospermic men, but 22.3.3.2 Testosterone undecanoate in tea seed oil
was better than that offered by condoms. While testosterone undecanoate was used as an oral
Even if these WHO studies represented a break- preparation solely in the West, in China it has been
through by confirming a principle of action (Waites marketed as an im preparation in tea seed oil for use
2003), they did not offer a practicable method. For a in hypogonadism (see Chapter 5), and has more
method requiring weekly intramuscular injections recently also been tested for male contraception. In a
is not acceptable for broad use. Moreover, several large multicenter efficacy study the first completed
months (on average four) were required before sperm since the WHO studies involving 308 Chinese men
production reached significant suppression. For this given monthly injections of 500 mg testosterone
reason research concentrated on the development undecanoate after a loading dose of 1000 mg, only
of long-acting testosterone preparations and on sub- 3% of the subjects did not suppress to azoospermia or
stances to improve overall effectiveness. severe oligozoospermia, and the remaining 97%
The WHO multicenter studies revealed an inter- induced no pregnancy (Gu et al. 2003).
esting phenomenon: the rate of azoospermia was The largest efficacy study to date was also per-
greater in East Asian than in Caucasian men (WHO formed in China, based on a loading dose of 1000
1990; 1996). This finding was also confirmed by inde- mg followed by monthly injections of 500 mg testos-
pendent studies using testosterone enanthate injec- terone undecanoate. In this, 898 men entered the
tions in men in Indonesia (Arysad 1993), Thailand efficacy phase during which only 9 pregnancies were
(Aribarg et al. 1996) and China (Cao et al. 1996). recorded. This represents a pregnancy rate of 1.1/100
person years (Gu et al. 2009). Thus, in China testos-
22.3.2 Testosterone buciclate terone undecanoate provides better protection against
Under the auspices of the WHO, a synthesis program pregnancy than condom use. Although injection
identified testosterone buciclate as a testosterone intervals of four weeks appeared to be an achievement
ester with long-lasting effectiveness. First tested in over the weekly injections of testosterone enanthate,
monkeys and then in hypogonadal patients, it showed the participants in a Chinese study considered the
a long effective phase of three to four months after a frequency of injections the most inconvenient part
single injection (see Chapter 15). A single injection of of this regimen (Zhang et al. 2006). Were testosterone
1200 mg in a contraceptive study resulted in suppres- undecanoate in castor oil also used in China this
sion of spermatogenesis comparable to that of weekly complaint could certainly be overcome.
enanthate injections (Behre et al. 1995). Unfortu-
22.3.3.3 Testosterone undecanoate in castor oil
nately, the WHO and the NIH, which jointly hold
the patent on testosterone buciclate, were unable to In Caucasian men im testosterone undecanoate has
find an industrial partner to further develop this not only been tested successfully for the treatment
promising ester for general use, so that its potential of male hypogonadism (see Chapters 14 and 15),
for male contraception has never been fully explored. but also in male contraception. Applying an improved
galenic preparation of testosterone undecanoate
(using castor oil instead of Chinese tea seed oil as
22.3.3 Testosterone undecanoate vehicle), injection intervals could be spaced to six
22.3.3.1 Oral testosterone undecanoate weeks, but, as with testosterone enanthate in weekly
If testosterone is suited for contraception, the orally injections, only two-thirds of the volunteers achieved
effective testosterone undecanoate should provide the azoospermia (Kamischke et al. 2000a). Although this
male contraceptive pill. This possibility was tested rate of azoospermia is not different from that

473
Chapter 22: Role of testosterone in male contraception

achieved with testosterone enanthate alone, the longer abdominal skin is compensated for by their low
injection interval represents a significant advantage. price and long duration of action. In further studies
A later pharmacokinetic study concluded that eight- testosterone pellets have only been used in combin-
week intervals of 1000 mg injections would be suffi- ation with other substances (see below).
cient for contraceptive purposes (Qoubaitary et al.
2006). Considering that 10- to 14-week intervals of 22.3.5 19-Nortestosterone
1000 mg testosterone undecanoate are required for
When searching for preparations with longer-lasting
substitution of hypogonadal men, about one-third
effectiveness, 19-nortestosterone-hexoxyphenylpropio-
more testosterone is required for contraception in
nate was tested, the spectrum of effects of which is very
normal volunteers.
similar to that of testosterone, and which has been used
Clinical studies to date have only included volunteers
as an anabolic since the 1960s (molecular structure is
with so-called normal semen values by WHO stand-
shown in Fig.22.1). This 19-nortestosterone ester
ards. As a male contraceptive should be applicable to all
injected every three weeks enabled azoospermia to be
interested men regardless of their semen parameters,
reached by as many men as by testosterone enanthate.
how volunteers with subnormal semen parameters
Thus the 19-nortestosterone ester is as effective as
respond to hormonal male contraception has recently
testosterone enanthate but allows a longer injection
been investigated (Nieschlag et al. 2011). During a 34-
interval (Schrmeyer et al. 1984; Knuth et al. 1985;
week treatment phase the volunteers received injections
Behre et al. 2001). However, 19-nortestosterone is not
of 1000 mg testosterone undecanoate in weeks 0, 6,
fully equivalent to testosterone as it is converted to
14 and 24. This was followed by a 24-week recovery
estrogens to a lesser degree than testosterone. Although
and follow-up period. Twenty-three men with normal
no side-effects were detected in the trials using 19-
semen parameters and 18 with sperm counts below 20
nortestosterone, long-term untoward effects, e.g. on
million completed the trial. The normal volunteers
bones, cannot be excluded. In the light of newer,
showed the expected response, with 17 suppressing
long-acting testosterone preparations, 19-nortestoster-
sperm counts below 1 million/ejaculate (13 showing
one appears less attractive for contraception.
azoospermia), and 6 not suppressing below 1 million
sperm/ejaculate. By the end of the recovery period all
sperm counts had returned to the range of starting 22.3.6 7a-Methyl-19-nortestosterone
values. The subnormal group showed a similar pattern Another synthetic androgen with possible application
with 13/18 ( 72%) men suppressing below 1 million/ in hypogonadism and in male contraception is 7a-
ejaculate (8/18 44% showing azoospermia) and the methyl-19-nortestosterone (MENT). It has been tested
remaining 5/18 ( 28%) not suppressing sperm counts in three doses of subdermal silastic implants in a multi-
below 1 million/ejaculate. All sperm counts returned to center study by the Population Council. In a dose-
the starting range. This demonstrated that, regarding dependent fashion azoospermia can be achieved at
suppressibility and reversibility, volunteers with normal the same rate as other testosterone preparations alone,
and subnormal sperm counts display the same pattern. i.e. in about two-thirds of the tested subjects; the
These results have a significant impact on the eligibility advantage being that the effect of one set of implants
of men for hormonal contraception, as those with sub- may last for as long as one year (von Eckardstein et al.
normal semen parameters would not need to be treated 2003). The potential of these implants alone or in
separately from those with normal parameters. combination with a gestagen is currently undergoing
testing by the Population Council (New York).

22.3.4 Testosterone pellets 22.4 Testosterone plus gonadotropin-


Pellets consisting of pure testosterone are used for
substitution in hypogonadism in some countries (see releasing hormone analogs
Chapter 15). In male contraceptive studies a one-time
application showed efficacy comparable to weekly
22.4.1 Testosterone plus
testosterone enanthate injections (Handelsman et al. gonadotropin-releasing hormone agonists
1992; McLachlan et al. 2000). The disadvantage In contrast to naturally occurring GnRH, GnRH
of minor surgery required for insertion under the agonists after producing an initial stimulation

474
Chapter 22: Role of testosterone in male contraception

CH3
CH3OH
Progesterone 20
C O Testosterone
CH3 CH3
CH3 16

CH3
3 5 O
4 6
O C O
CH3
OCOCH3
CH3
17a-Hydroxy- CH3
progesterone acetate

O C O
CH3
OCOCH3
CH3

Medroxyprogesterone
acetate 6 CH3
O
CH3
C O
CH3
OCOCH3
H2C CH3
1
Cyproterone 2
acetate 6
O

Cl
Fig. 22.1 Progestins derived from 17-hydroxyprogesterone tested in hormonal male contraception.

of gonadotropin release for approximately two Altogether, GnRH agonists in combination with
weeks lead to GnRH receptor downregulation and testosterone did not prove useful in male contra-
thereby to suppression of LH and FSH synthesis and ception. At times it has been suggested that higher
secretion. doses of the GnRH agonists should be used, but
Between 1979 and 1992, 12 trials for hormonal currently no further clinical studies appear to be
male contraception using GnRH agonists, mostly in under way.
combination with testosterone, were published (for a
review see Nieschlag et al. 1992). Altogether 106 vol-
unteers participated in these trials. The GnRH agon-
22.4.2 Testosterone plus gonadotropin-
ists Decapeptyl (triptorelin acetate), buserelin and releasing hormone antagonists
nafarelin were administered at daily doses of 5500 In contrast to GnRH agonists, GnRH antagonists
mg/volunteer for periods of 1030 weeks. In about produce a precipitous and prolonged fall of LH and
30% of men, sperm production could be suppressed FSH serum levels in men (e.g. Pavlou et al. 1989;
below 5  106/ml, and azoospermia occurred in 21 Behre et al. 1994). It took much longer to develop
men; while in the remaining volunteers, sperm GnRH antagonists that were suitable for clinical
numbers were only slightly reduced or remained application than it did for GnRH agonists, and clin-
unaffected. One explanation for the ineffectiveness ical trials using GnRH antagonists for male contra-
of GnRH agonist plus androgen is the escape of FSH ception started some 12 years later than those using
suppression after several weeks of GnRH agonist agonists. To date, results have become available from
treatment (Behre et al. 1992; Bhasin et al. 1994). five clinical trials using GnRH antagonists for male

475
Chapter 22: Role of testosterone in male contraception

contraception (for review see Nieschlag and Behre 22.5 Testosterone combined
1996; Swerdloff et al. 1998; Behre et al. 2001).
Overall, 35 of the 40 volunteers (88%) who took with progestins
part in these studies became azoospermic, most within The potency of gestagens to suppress gonadotropins
three months. This is a much better rate of complete is well known, and it was therefore obvious to com-
suppression than that produced by the administration bine testosterone with gestagens to achieve higher
of testosterone enanthate alone. Although studies in contraceptive efficacy. An overview of all studies
monkeys had suggested that delayed testosterone using testosterone in combination with a progestin
administration would increase the effectiveness of performed to date is given in Table 22.1, and the
GnRH antagonists (Weinbauer et al. 1987; 1989), in gestagen formulae are shown in Figs. 22.1 and 22.2.
men GnRH administration followed by delayed testos- Unfortunately, a systematic comparison of the
terone administration (azoospermia in 20/22 men) different gestagens with regard to their contraceptive
offered little advantage over concomitant GnRH and potency in males has never been performed. Even
testosterone administration (azoospermia in 15/18 worse, a Cochrane Review analyzing 45 clinical trials
men). It should also be noted that, in the later studies came to the conclusion that the studies comprised
with concomitant administration, all 14 volunteers too few volunteers, so that significant differences
became azoospermic (Pavlou et al. 1994; Behre et al. between the various steroid combinations could not
2001). The major advantage of using GnRH antagon- be detected (Grimes et al. 2007). In the following,
ists is the short time required to achieve azoospermia; important studies are briefly summarized to highlight
that is, within 68 weeks, which is considerably the cumbersome and often frustrating development
shorter than the mean of 17 weeks required in of effective male contraception (Nieschlag 2009).
Caucasian men when testosterone alone was used
(WHO 1995).
These results are promising. However, the antag- 22.5.1 Testosterone or
onists and regimen tested so far require a daily 19-nortestosterone plus depot
injection, which makes them unacceptable for con-
traceptive purposes. Recently, modern GnRH antag- medroxyprogesterone acetate
onists for treatment of hormone-dependent prostate 19-Norethisterone (norethindrone), medroxyproges-
cancer became available that have to be injected only terone acetate (MPA), depot-MPA (DMPA), 17-
on a monthly basis (Boccon-Gibod et al. 2011). hydroxyprogesterone capronate and megestrol acetate
In addition, orally effective GnRH antagonists have were used in early clinical trials initiated by the WHO
been shown to suppress gonadotropins in normal (19721995) and the Population Council (Schearer
men (Amory et al. 2009). The combination of these et al. 1978) (molecular structures are in Figs.22.1
new GnRH antagonists with testosterone has the and 22.2). The most favorable combination was the
clear potential for effective hormonal contraception monthly im injection of 200 mg DMPA plus 200 mg
and should be tested in appropriate clinical trials in testosterone enanthate or testosterone cypionate; this
the near future. combination gave the best results in suppressing
It could be argued that the high price of GnRH spermatogenesis, and the incidence of untoward
antagonists may preclude their development as male side-effects was low. Twelve volunteers were injected
contraceptives, which need to be affordable and in weekly with 200 mg 19-nortestosterone hexoxyphe-
the same price range as comparable female methods. nylpropionate, followed by injections with the same
In order to shorten the period of use of GnRH dose every 3 weeks up to week 15. In addition, the
antagonists, two studies have investigated the possi- volunteers were injected with 250 mg DMPA in weeks
bility of applying GnRH antagonists only in an ini- 0, 6 and 9. Azoospermia was achieved in 9 of 12
tial suppression phase, and then continuing with the volunteers during the study course, while, in 3 of the
androgen alone (Swerdloff et al. 1998; Behre et al. remaining 4 volunteers, spermatogenesis was sup-
2001). Although successful in the monkey model pressed to single sperm, and, in one volunteer, to a
(Weinbauer et al. 1994), studies in men produced sperm concentration of 1.3 million/ml.
contradictory results, so that this approach would The promising results prompted the WHO Task
require further experimentation. Force on Methods for the Regulation of Male Fertility

476
Table 22.1 Overview of studies on hormonal male contraception using either testosterone alone or in combination with progestins

Reference Number Ethnic Androgen dose Progestin Azoospermia Severe oligozoospermia Oligozoospermia
of origin dose (n) below 1 million/ml (n) below 3 million/ml
subjects (n)
Testosterone alone
WHO 1990 271 Mixed TE 200 mg im/week None 157 ?? ??
Behre et al. 8 Caucasian TB 1000 mg im once None 3
1995
Handelsman 9 Unknown T pellets 1200 mg None 5 4 0
et al. 1992
Handelsman 10 Unknown T pellets 400 mg None 0 0 0
et al. 1996
Handelsman 10 Unknown T pellets 800 mg None 4 0 0
et al. 1996
Meriggiola 5 Caucasian TE 100 mg im/week None 5 0 0
et al. 1996
Bebb et al. 18 Caucasian TE 100 mg im/week None 6 4 1
1996
WHO 1996 225 Mixed TE 200 mg im/week None 157 29 8
Zhang et al. 12 Chinese TU* 500 mg im/4 weeks None 11 1 0
1999
Zhang et al. 12 Chinese TU* 1000 mg im/4 weeks None 12 0 0
1999
Kamischke 14 Caucasian TU 1000 mg im/6 weeks None 7 4 1
et al. 2000a
McLachlan 5 Not TE 200 mg im/week None 4 men 0.1 or 4 0
et al. 2002 stated azoospermia
von 35 Caucasian MENT implants, 3 doses None 10 3
Eckardstein
et al. 2003
Gu et al. 2003 305 Chinese TU* 500 mg im/4 weeks None 284 6 6
Gu et al. 2009 898 Chinese TU* 500 mg im/4 weeks None 855 43
Nieschlag 41 Caucasian TU 1000 mg im/ None 25 9
et al. 2011 6, 14 and 24 weeks
Depot medroxyprogesterone acetate (DMPA)
Alvarez-Sanchez et al. 8 Dominican TE 250 mg im/week DMPA 150 mg/4 weeks 4 3 1
1977 republic
Alvarez-Sanchez et al. 10 Dominican TE 250 mg im/week DMPA 300 mg/4 weeks 7 2 0
1977 republic
Brenner et al. 1977 6 Caucasian TE 200 mg im/week DMPA 100 mg/4 weeks 1 2 1
Brenner et al. 1977 3 Caucasian TE 200 mg im/week DMPA 150 mg/4 weeks 1 0 0
Frick et al. 1977 12 Caucasian TE 250 mg im/week DMPA 100 mg im/4 weeks 6 4 0
Frick et al. 1977 6 Caucasian T-propionate 4 rods DMPA 100 mg im/4 weeks 2 0 0
Melo and Coutinho 1977 11 Brazilian TE 200 mg im/week DMPA 100150 mg 11 men 0.1 or azoospermia 0 ??
im/4 weeks
Faundes et al. 1981 10 Dominican TE 500 mg im/week DMPA 150 mg/4 weeks 8 1 0
republic
Frick et al. 1982 4 Caucasian TE 500 mg/4 weeks 150 mg/4 weeks 4 0 0
Frick et al. 1982 5 Caucasian TE 250 mg/2 weeks 75 mg/2 weeks 5 0 0
WHO 1993 45 Indonesian 19-Nortestosterone 200 DMPA 250 mg im/6 weeks 44 1 0
mg
im/3 weeks
WHO 1993 45 Indonesian TE 200 mg im/3 weeks DMPA 250 mg im/6 weeks 43 2 0
Knuth et al. 1989 12 Caucasian 19-Nortestosterone 200 DMPA 250 mg im/6 weeks 6 4 2
mg
im/3 weeks
Wu and Aitken 1989 10 Caucasian TE 250 mg im/week DMPA 200 mg/4 weeks 6 0 4
Pangkahila 1991 10 Indonesian TE 100 mg im/week DMPA 100 mg/4 weeks 10 0 0
Pangkahila 1991 10 Indonesian TE 250 mg im/week DMPA 200 mg/4 weeks 10 0 0
Handelsman et al. 1996 10 Not stated T pellets 800 mg DMPA once 300 mg im 9 0 1
McLachlan et al. 2002 5 Not stated TE 200 mg im/week DMPA once 300 mg im 5 men 0.1 or azoospermia 5 0
Turner et al. 2003 53 Unknown T pellets 800 mg/16 weeks DMPA 300 mg im/12 weeks 49 2 0
Gu et al.2004 30 Chinese TU* 1000 mg/8 weeks DMPA 150 or 300 mg/8 weeks 28 1 1
Page et al. 2006 38 Not stated T gel 100 mg/day DMPA 300 mg/12 weeks 31 2 3
GnRH antagonist
Levonorgestrel (LNG)
Fogh et al. 1980 5 Caucasian TE 200 mg/4 weeks LNG 250 g po/day 1 ?? 1
Fogh et al. 1980 5 Caucasian TE 200 mg im/4 weeks LNG 500 g po/day 2 ?? ??
Bebb et al. 1996 18 Caucasian TE 100 mg im/week LNG 500 g po/day 12 2 3
Anawalt et al. 1999 18 Caucasian TE 100 mg im/week LNG 125 g po/day 11 5 1
Anawalt et al. 1999 18 Caucasian TE 100 mg im/week LNG 250 g po/day 14 2 0
Ersheng et al. 1999 16 Chinese TU 250 mg im/4 weeks Sino-Implant 2 rods 6 0 1
Kamischke et al. 2000a 14 Caucasian TU 1000 mg im/6 weeks LNG 250 g po/day 8 4 2
Gaw Gonzalo et al. 2002 20 Mixed Testoderm TTS 2 patches/day Norplant II 4 rods 7 5 2
Gaw Gonzalo et al. 2002 15 Mixed Testoderm TTS 2 patches/day LNG 125 g po/day 5 1 1
Gaw Gonzalo et al. 2002 14 Mixed TE 100 mg im/week Norplant II 4 rods 13 1 0
Pllnen et al. 2001 5 Caucasian DHT gel 250 mg/day LNG 30 g po/day 0 0 1
Pllnen et al. 2001 5 Caucasian DHT gel 250 mg/day Jardelle (LNG) 1 rod 0 0 0
Pllnen et al. 2001 8 Caucasian DHT gel 500 mg/day Jardelle (LNG) 2 rods 0 0 0
Pllnen et al. 2001 7 Caucasian DHT gel 250 mg/day Jardelle (LNG) 4 rods 0 0 0
Gui et al.2004 41 Chinese TU* 500 or 1000 mg/8 weeks LNG 4 implants 31 5 4
Anawalt et al.2005 41 Mixed TE 100 mg/week LNG 31 g or 62 g/day 25 13 2
Wang et al. 2006 19 Caucasian Chinese T implants/1518 weeks LNG 4 implants 13
21 19
Wang et al. 2007 18 Chinese TU* 500 mg/6 weeks LNG 250 mg po/day 17 1
Norethisterone enanthate (NETE)
Kamischke et al. 2001 14 Caucasian TU 1000 mg im/6 weeks NETE 200 mg/6 weeks 13 0 0
Kamischke et al. 2002 14 Caucasian TU 1000 mg im/6 weeks NETE 200 mg/6 weeks 13 1 0
Kamischke et al. 2002 14 Caucasian TU 1000 mg im/6 weeks NETE 400 mg/6 weeks 13 1 0
Kamischke et al. 2002 14 Caucasian TU 1000 mg im/6 weeks NETA 10 mg po/day 12 2 0
Meriggiola et al. 2005 10 Caucasian TU 1000 mg/8 weeks NETE 200 mg/6 weeks 9
8 Caucasian TU 1000 mg/12 weeks NETE 200 mg/12 weeks 3
Qoubaitary et al. 2006 10 Mixed TU 750 mg/8 weeks NETE 250 mg/8 weeks 5 2 1
10 Mixed TU 1000 mg/8 weeks NETE 250 mg/8 weeks 10
CONRAD 2011 Mixed TU 1000 mg/8 weeks NETE 200 mg/8 weeks
Cyproterone acetate (CPA)
Meriggiola et al. 1996 5 Caucasian TE 100 mg im/week CPA 50 mg po/day 3 0 1
Meriggiola et al. 1996 5 Caucasian TE 100 mg im/week CPA 100 mg po/day 5 0 0
Meriggiola et al. 1998 5 Caucasian TE 100 mg im/week CPA 12.5 mg po/day 3 2 0
Meriggiola et al. 1998 5 Caucasian TE 100 mg im/week CPA 25 mg po/day 5 0 0
Meriggiola et al. 2002a 9 Caucasian TE 100 mg im/week CPA 5 mg po/day 6 3 0
Meriggiola et al. 2002a 7 Caucasian TE 200 mg im/week CPA 5 mg po/day 0 4 2
Meriggiola et al. 2003 24 Caucasian TU 1000 mg/6 weeks CPA 20 and 2 mg po/day 13 11
Desogestrel (DSG) or etonogestrel (ENG)
Wu et al. 1999 8 Caucasian TE 50 mg im/week DSG 300 g po/day 8 0 0
Wu et al. 1999 7 Caucasian TE 100 mg im/week DSG 150 g po/day 4 3 0
Wu et al. 1999 8 Caucasian TE 100 mg im/week DSG 300 g po/day 6 0 1
Anawalt et al. 2000 7 Caucasian TE 50 mg im/week DSG 150 g po/day 4 1 0
Anawalt et al. 2000 8 Caucasian TE 100 mg im/week DSG 150 g po/day 8 0 0
Anawalt et al. 2000 8 Caucasian TE 100 mg im/week DSG 300 g po/day 7 1 0
Kinniburgh et al. 2001 8 Caucasian T pellets 400 mg/12 weeks DSG 150 g po/day 6 2 0
Kinniburgh et al. 2001 7 Caucasian T pellets 400 mg/12 weeks DSG 150 g po/day 5 1 0
Anderson et al. 2002a 9 Black T pellets 400 mg/12 weeks DSG 150 g po/day 9 0 0
Anderson et al. 2002a 11 Mixed T pellets 400 mg/12 weeks DSG 150 g po/day 9 0 1
Anderson et al. 2002a 8 Black T pellets 400 mg/12 weeks DSG 300 g po/day 8 0 0
Anderson et al. 2002a 12 Mixed T pellets 400 mg/12 weeks DSG 300 g po/day 8 0 0
Anderson et al. 2002b 14 Caucasian T pellets 400 mg/12 weeks Implanon (ENG) 1 rod 9 1 3
Anderson et al. 2002b 14 Caucasian T pellets 400 mg/12 weeks Implanon (ENG) 2 rods 9 4 0
Kinniburgh et al. 2002 15 Caucasian T pellets 400 mg/12 weeks DSG 300 g po/day 15 0 0
Kinniburgh et al. 2002 18 Chinese T pellets 400 mg/12 weeks DSG 300 g po/day 18 0 0
Kinniburgh et al. 2002 18 Chinese T pellets 400 mg/12 weeks DSG 150 g po/day 11 2 2
Kinniburgh et al. 2002 13 Caucasian T pellets 400 mg/12 weeks DSG 150 g po/day 11 2 0
Brady et al.2004 9 Not stated T pellets 400 mg/12 weeks Etonogestrel implants 9 2
Walton et al. 2007 16 Caucasian T pellets 600 mg/12 weeks Etonogestrel implants 11 2
10 Caucasian MENT implants Etonogestrel implants 3 5
Mommers et al. 2008 134 Caucasian TU 750 mg/12 weeks Etonogestrel implants high dose 125
112 Caucasian TU 750 mg/10 weeks Etonogestrel implants low dose 100
TU 1000 mg/12 weeks
Self-applicable
Nieschlag et al. 1978 7 Caucasian Andriol 240 mg po/day None 1 0 0
Guerin and Rollet 1988 13 Caucasian Andriol 160 mg po/day NETA 10 mg po/day 7 2 3
Guerin and Rollet 1988 5 Caucasian T gel 250 mg/day NETA 5 mg po/day 4 1 0
Guerin and Rollet 1988 5 Caucasian T gel 250 mg/day NETA 10 mg po/day 5 0 0
Guerin and Rollet 1988 8 Caucasian T gel 250 mg/day MPA 20 mg po/day 5 0 1
Meriggiola et al. 1997 8 Caucasian Andriol 80 mg po/day CPA 12.5 mg po/day 1 3 2
Hair et al. 1999 4 Caucasian Andropatch 2 patches/day DSG 75 g po/day 0 1 0
Hair et al. 1999 6 Caucasian Andropatch 2 patches/day DSG 150 g po/day 3 0 0
Hair et al. 1999 7 Caucasian Andropatch 2 patches/day DSG 300 g po/day 4 1 0
Bchter et al. 1999 12 Caucasian Testoderm TTS 2 patches/day LNG 250 g po, later 500 g 2 3 0
Gaw Gonzalo et al. 2002 19 Mixed Testoderm TTS 2 patches/day None 5 0 1
Pllnen et al. 2001 2 Caucasian DHT gel 250 mg/day None 0 0 0
Soufir et al. 2011 35 Not stated T gel 100125 mg/day MPA 2 10 mg daily 23 5 1
Source: updated from Kamischke and Nieschlag (2004).
Abbreviations: T, testosterone; TE, testosterone enanthate; TB, testosterone buciclate; TU*, testosterone undecanoate in tea seed oil; TU, testosterone undecanoate in castor oil; MENT,
7-methyl-19-nortestosterone; NETA, norethisterone acetate; po, per os; ??, not known, can not be extracted from the paper.
Chapter 22: Role of testosterone in male contraception

Testosterone CH3OH OH
CH3
CH2CN
CH3

Dienogest
O O

OH Desogestrel Etonogestrel
19-Nortestosterone CH3
OH H3C OH
H 3C CH2
H2C C OH
H2C C CH

O O

19-Norethisterone OH OH
CH3 H 3C CH2
C HC C HC

O O Levonorgestrel

Fig. 22.2 Progestins derived from 19-nortestosterone tested in hormonal male contraception.

to launch a large-scale multicenter trial in five centers given alone in weekly im injections or in combin-
in Indonesia, comparing the effectiveness of testoster- ation with an injection of 300 mg DMPA showed
one enanthate, or 19-nortestosterone hexyoxyphenyl- that the suppression rate was not greater when
propionate, in combination with DMPA (WHO DMPA was added (McLachlan et al. 2002). How-
1993). Surprisingly, 43/45 and 44/45 subjects in the ever, when subcutaneous testosterone implants of
testosterone and the 19-nortestosterone groups 200 mg were applied every four or six months in
respectively suppressed to azoospermia. Unfortu- combination with 300 mg DMPA given every three
nately, this study had failed to include groups treated months, 53/54 men achieved azoospermia or sup-
with the androgens alone, so that it remained unclear pression below 1  106 sperm/ml. During a 12-
whether the azoospermia rates of 97% and 98% were month efficacy phase with otherwise unprotected
due to the combined treatment or could also be intercourse, no pregnancy occurred (35.5 person
achieved by the androgens alone. years) (Turner et al. 2003). The differences between
The latter possibility appears likely in the light of the studies highlight the fact that obviously the
ethnic differences between Caucasian and East Asian kinetics of testosterone are very important, since
men. Although ultimately effective, the disadvantage the implants produce very stable serum levels and
remains that it took almost 20 weeks to reach azoo- the testosterone enanthate injections cause high
spermia or the lowest sperm counts in these volun- peaks and troughs. Although the combination of
teers. Thus, more rapid onset of sperm suppression is an implant with an injection every three months
required. may not be ideal, this study was the first to demon-
A more recent study testing contraceptive effi- strate the contraceptive efficacy of a testosterone
cacy and using either 200 mg testosterone enanthate plus progestin combination.

483
Chapter 22: Role of testosterone in male contraception

In order to test whether one of the two steroid again emphasizes the need for steady serum testoster-
entities could be self-administered, the addition of one levels to suppress gonadotropins, even when co-
a testosterone transdermal gel to the DMPA injec- administered with a potent gestagen.
tions (300 mg/three months) was tested (Page et al. Similarly it was shown that the combination of 0.5
2006). The results were comparable to those from mg levonorgestrel given orally with transdermal DHT
trials where DMPA was combined with injectable was quite ineffective; nor did the combination of
testosterone. transdermal DHT with levonorgestrel implants lead
In order to develop a self-applicable male hormo- to sufficient suppression of spermatogenesis (Pll-
nal contraceptive, Soufir et al. (2011) initiated an nen et al. 2001).
efficacy study combining transdermal testosterone When the long-acting testosterone preparation
(100125 mg daily) with oral MPA (2  10 mg daily). testosterone undecanoate (in castor oil) given at six-
Of the 35 eligible couples, 25 entered the efficacy week intervals was combined with oral levonorgestrel,
phase, and all female partners together were exposed the progestin did not enhance the effect of testoster-
for 211 months to otherwise unprotected intercourse. one undecanoate alone (Kamischke et al. 2000b).
The one pregnancy occurred in a couple that showed However, when levonorgestrel was administered in
bad compliance. Numbers of participants and months four capsules delivering about 160 mg levonorgestrel
of exposure are too low for statistical analysis, but the (Norplant II = Jadelle)/per day together with weekly
results show that a self-applicable male hormonal injections of 100 mg testosterone enanthate, 93% of
contraceptive is possible. the subjects achieved azoospermia and all suppressed
Recovery to baseline semen parameters appears to to oligozoospermia below 1  106/ml sperm (Gaw
be rather slow in studies employing DMPA. This may Gonzalo et al. 2002).
be due to secondary depots of this progestin formed When MENT implants were combined with levo-
in the subcutaneous and abdominal fat, and requires norgestrel implants in different doses, a clear dose-
special attention should studies be extended over dependent effect could be observed, but it remains
several years. undetermined whether implants with sufficiently long
duration can be manufactured. Non-biodegradable
implants that have to be removed surgically from
22.5.2 Testosterone plus levonorgestrel the implantation site when contraceptive protection
Levonorgestrel has been widely used for contracep- is no longer required appear impractical for wide-
tion in females, either orally or as an implant, and spread use unless they can be left in situ for long
has proved safe and effective. Although early studies periods (Wang et al. publication in preparation).
combining 0.5 mg levonorgestrel given orally with
testosterone enanthate were not very encouraging
(Fogh et al. 1980), later trials comparing testoster-
22.5.3 Testosterone plus cyproterone
one enanthate (100 mg/week) alone with testoster- acetate
one enanthate in combination with 0.5 mg Animal studies and studies in sexual delinquents have
levonorgestrel given orally showed that the combin- shown that the antiandrogen cyproterone acetate,
ation resulted in more pronounced suppression of which can be considered a potent progestin, sup-
spermatogenesis than testosterone enanthate alone presses spermatogenesis, an effect exerted through
(Bebb et al. 1996). suppression of pituitary gonadotropin secretion. In
Encouraged by the renewed interest in levonor- clinical trials using 5 to 20 mg cyproterone acetate
gestrel, we conducted a self-administration trial com- per day for up to 16 weeks, sperm counts and motility
bining oral levonorgestrel with a transdermal were reduced markedly (Fogh et al. 1979; Moltz et al.
testosterone patch applied to the trunk (Bchter 1980; Wang and Yeung 1980). Thus, cyproterone
et al. 1999). Unfortunately the results were disap- acetate appeared to be a possibility for male fertility
pointing, as suppression of spermatogenesis was control. However, decreases in serum testosterone
insufficient. We presume that the testosterone dose levels to below normal were also observed. Some of
absorbed from the transdermal systems was too low the volunteers complained of fatigue, lassitude and
and often impeded by inadequate adhesiveness to the decrease in libido and potency attributable to the
skin of the systems (Bchter et al. 1999). The study diminished testosterone levels.

484
Chapter 22: Role of testosterone in male contraception

When, later, cyproterone acetate was combined While the group receiving 50 mg testosterone
with testosterone enanthate injections at even enanthate showed complete suppression of spermato-
higher doses of 50 and 100 mg, it effectively sup- genesis, i.e. azoospermia, the other groups achieved
pressed spermatogenesis (Meriggiola et al. 1996), only incomplete suppression. In the most effective
but even when lower doses of cyproterone acetate group, total serum testosterone levels were found in
were administered, antiandrogenic effects pre- the range of the lower limit of normal men, and this
vailed and the volunteers showed decreased red may explain why the volunteers complained of
blood, preventing this antiandrogenic gestagen decreased sex drive, depression, fatigue and nocturnal
from being an attractive combination for male sweating (Wu et al. 1999).
contraception (Meriggiola et al. 1998). Although In a two-center study in Edinburgh and Shang-
the attempt to create a male pill by co-adminis- hai, testosterone pellets (400 mg every three months)
tration of oral testosterone undecanoate with oral were combined with either 150 or 300 mg desogestrel/
cyproterone acetate led to suppression of sperm- day orally (Kinniburgh et al. 2001). Azoospermia
atogenesis, it had to be discontinued because of a was achieved in all 28/28 men receiving the 300 mg
decrease in hemoglobin and hematocrit caused by desogestrel dose. Disregarding the fact that a com-
the antiandrogen (Meriggiola et al. 1997). How- bination of an implant with an oral pill might not
ever, when combining 1000 mg testosterone unde- offer a highly attractive option, these results are
canoate every six weeks with 20 mg CPA daily quite promising.
initially, followed by only 2 mg CPA/day, the This group continued their investigations using
initial suppression of spermatogenesis could be etonogestrel, the active metabolite of orally active
maintained and antiandrogenic effects prevented desogestrel, as an implant which was recently
(Meriggiola et al. 2003). licensed for use as a female contraceptive (Impla-
non). Twenty-eight men received one or two etono-
gestrel implants which provide contraceptive
22.5.4 Testosterone plus dienogest protection in females for three years, but the
The latest progestin to be tested for male contracep- implants were removed from the volunteers after
tive purposes is the orally effective dienogest. This six months. In addition, they received 400 mg tes-
is another 19-norprogestin in which position 17 is tosterone pellets at the beginning of the study and
not substituted by the common ethinyl group, but after three months. Nine men in each group
by a cyanomethyl group and a double bond is achieved azoospermia and, in the group with two
introduced in ring B. When given at 2, 5 or 10 implants, sperm counts fell to 0.1  106/ml in 13/
mg doses over 21 days, 10 mg resulted in a suppres- 14 men (Anderson et al. 2002b).
sion of gonadotropins comparable to 10 mg of In the first (and so far last) industry-sponsored
cyproterone acetate. Semen parameters were not trial, Organon and Schering decided to test etono-
affected, as one would expect with this short appli- gestrel implants with testosterone undecanoate
cation period (Meriggiola et al. 2002b). As dieno- injections in various combinations (Mommers
gest displays only mild antiandrogenic activity, this et al. 2008). This study involved 354 volunteers in
substance may be a possible candidate for future seven treatment groups receiving either placebo or
trials. However, to date further testing has only 7501000 mg testosterone undecanoate every 1012
been performed in rats (Misro et al. 2009). weeks with two doses of etonogestrel, for 4244
weeks. Ninety percent of treated men suppressed
22.5.5 Testosterone plus desogestrel or spermatogenesis to 1 million/ml ejaculate.
Although the combination of an implant with injec-
etonogestrel tions may not appear too attractive for practical use,
Orally administered desogestrel, a levonorgestrel the study had a high success rate and could have
derivative, was evaluated in clinical trials using formed the basis for a phase III efficacy study.
300 mg/day combined with weekly injections of 50 or Unfortunately, both companies discontinued their
100 mg testosterone enanthate for 24 weeks. A third male contraception programs when they were taken
group received 150 mg/day desogestrel and 100 mg over by other firms who were at that stage not
testosterone enanthate per week intramuscularly. interested in male contraception.

485
Chapter 22: Role of testosterone in male contraception

22.5.6 Testosterone plus possibly or probably related to the study regimen.


By the time of the suspension of the study (April 5,
19-norethisterone 2011) 321 men had been enrolled into the study, 100
19-norethisterone, one of the earliest progestins men had completed the 12-months efficacy phase
derived from testosterone (Djerassi et al. 1954), is and 103 men were still in the suppression or efficacy
characterized by some undesirable androgenicity phase. All subjects continued to be followed until the
when given to women, which might be of advantage end of the recovery phase (CONRAD 2011). Thus,
when administered to men. An early study with only a this largest efficacy trial ever performed worldwide
few volunteers using a combination of orally effective came to an unhappy end.
19-norethisterone acetate with either a transdermal
testosterone gel or oral testosterone undecanoate led
to azoospermia in all volunteers (Guerin and Rollet
22.6 Side-effects of hormonal male
1988). Considering its properties and these promising contraception
results, it was surprising that it took another 10 years Possible side-effects of hormonal male contraception
to investigate the use of 19-norethisterone more might be caused by too high or too low testosterone
systematically. levels or by additional substances. Decreased testicu-
In a pharmacokinetic study, single injections of lar volumes reflecting suppression of spermatogenesis
200 mg 19-norethisterone enanthate led to a marked are inherent to all hormonal methods, but not con-
suppression of the gonadotropins (FSH for 29 days), sidered a serious effect by the volunteers as long as
testosterone, SHBG and sperm (Kamischke et al. sexual function remains unaltered. In all major stud-
2000b). When testosterone undecanoate became ies performed to date, sperm counts returned to
available in the form of im depot injections, it was normal levels (Liu et al. 2006). Weight gain is most
combined with norethisterone enanthate, and volun- likely an anabolic effect of testosterone. Due to the
teers achieved azoospermia or severe oligozoospermia high peak serum testosterone levels caused by testos-
in all but one. The additive effect to testosterone terone enanthate in the earlier studies, acne and mild
undecanoate alone was striking. An injected dose of gynecomastia could be observed in individual cases.
200 mg 19-norethisterone enanthate every six weeks Except for local skin reactions, side-effects of GnRH
was as effective as 400 mg, so that 200 mg appears to analogs are mainly attributable to decreased testoster-
be a useful dose. Although 19-norethisterone acetate one levels, not sufficiently compensated for by testos-
10 mg given orally daily in combination with im terone supplementation. Sweating and, in particular,
testosterone undecanoate is as effective as injected nocturnal sweating is a feature of some added
norethisterone enanthate, the combination of the progestins.
two steroids in one injection appears quite attractive Depending on the type and doses of progestin,
(Kamischke et al. 2000a; 2001; 2002). In an ensuing significant decreases are observed in SHBG. This
dose-finding study, Meriggiola et al. (2005) showed indicates the influence of progestins on liver function
that it might be possible to extend the injection inter- and may enhance the androgenicity of the testoster-
vals to eight weeks. one preparation, since the unbound free fraction of
Based on these findings, the WHO, together with testosterone in circulation may increase. Some of the
CONRAD, initiated a large-scale efficacy trial effects seen when progestins are added may be due to
intended to include 440 couples in 11 centers world- this phenomenon. When adding levonorgestrel or 19-
wide. The men received 1000 mg testosterone unde- norethisterone acetate or enanthate an increase in
canoate plus 200 mg norethisterone enanthate im prolactin is seen, which remains without biological
every eight weeks. Although contraceptive efficacy significance. An increase in red blood was more pro-
appeared to be satisfactory, the study was prema- nounced when progestins were added to testosterone
turely suspended because of undesirable side-effects, than when testosterone was given alone. Hemostasis
especially depression and other mood changes, is affected by testosterone alone (downregulation) and
increase of sexual desire and pain at the injection by progestin (in this case norethisterone) alone (upre-
site. These side-effects were not unexpected, but gulation), but given in combination, the effects appear
occurred more often that expected. Two serious to be neutralized (Zitzmann et al. 2002). In the sus-
adverse events (SAEs) were judged to be either pended large WHO/CONRAD study, depression and

486
Chapter 22: Role of testosterone in male contraception

other mood changes were the major complaints and it Development, some medical research councils and a
remains to be seen whether this was due to too little few foundations, male contraception would long have
testosterone or whether this was a genuine effect of been abandoned (Nieschlag 2009). The principle and
norethisterone. effectiveness of hormonal male contraception has
been demonstrated in many studies. The fact that
the majority of clinical trials on hormonal male con-
22.7 Acceptability of hormonal male traception have been published in high-ranking jour-
contraception nals emphasizes the high priority the scientific
The acceptability of hormonal male contraception can community attributes to these endeavors. Investiga-
definitely only be assessed when a final product tors are so convinced of the validity of the concept of
becomes available. Nevertheless, it should be remem- hormonal male contraception that they drafted rec-
bered that worldwide about 15% of all couples prac- ommendations for regulatory approval for male hor-
ticing contraception rely on male methods, albeit with monal contraception at their annual summit meetings
varying preferences, and the proportion of men prac- (since 1997) (Nieschlag and 10th Summit Meeting
ticing contraception is increasing. Thus in the Nether- Group 2007). Little more would have been required
lands the percentage of vasectomized men whose to convince industry to bring this development to
wives were of reproductive age rose from 2 to 10.5% fruition. However, the recent suspension of the
from 1975 to 2008 and from 8 to 12.2% in the USA; WHO/CONRAD study implies an unexpected set-
the highest rates of vasectomized men are found in back in the entire development of hormonal male
the United Kingdom and in New Zealand. World- contraception. Unless clear mistakes in the design or
wide, however, only 2.7% of men are vasectomized. the conduct of the study can be detected during the
Similarly, the use of condoms for contraception varies current analysis of the study, it will be very difficult to
from country to country with a worldwide average of get the pharma industry back on track or to attract
5.7%. It is to be expected that the percentage of men new funding.
willing to practice contraception varies between cul- On a more general note, comparing the situation
tures and with methods available. According to a with the development of the female pill, the lack of
survey in Hong Kong and Shanghai some years ago, public advocacy for male contraception is striking.
half the men interviewed were willing to take a daily Male contraception lacks prominent advocates, as
contraceptive pill; in Edinburgh and Cape Town two- the development of female contraception benefited
thirds were willing to do so (Anderson and Baird from personalities such as Margaret Sanger (1879
1997; Martin et al. 2000). Also acceptability in Aus- 1966) and Katherine McCormick (18751967). Hor-
tralian men was found to be high (Weston et al. monal male contraception requires similar advocacy
2002). After more than 50 years of female oral con- to finally result in a marketable product (Nieschlag
traception, the attitude of men towards new methods 2009). However, it should not be forgotten that there
of male contraception has changed. Worldwide is a fundamental sex difference in the perception of
surveys showed men to be willing to use pharmaco- contraception. For women contraception means per-
logical contraceptive methods (Heinemann et al. sonally avoiding pregnancy with all of its medical,
2005). In addition, despite commonly expressed social and economic implications including the threat
views to the contrary, most women would trust their of death. In contrast, for men contraception is more
male partners to use a hormonal method (Glasier of an intellectual issue regarding respect for the part-
et al. 2000; Glasier 2010). ner and avoiding financial, social and legal obligations
of fatherhood. Without emotion, however, it remains
difficult to attract protagonists campaigning for male
22.8 Outlook contraception.
While a large proportion of clinical research is driven
by the pharmaceutical industry, in the case of male 22.9 Key messages
contraception industry fails. Without the long-range  Testosterone-induced azoospermia leads to
perspective and endurance of institutions and organ- effective, safe and reversible male contraception.
izations such as the WHO, Population Council, the Suppression of spermatogenesis to below
National Institute of Child Health and Human

487
Chapter 22: Role of testosterone in male contraception

1 million/ml sperm may still be compatible with modalities (oral or transdermal) require a high
protection from pregnancy. rate of compliance.
 About two-thirds of Caucasian and almost all East  Side-effects of hormonal contraception were rare
Asian men reach azoospermia when given and tolerable until the WHO/CONRAD study was
weekly testosterone enanthate injections or prematurely suspended because of mood
four- to six-weekly injections of testosterone alterations, the reasons for which have to be
undecanoate. clarified.
 In order to speed up suppression of  Acceptability of a hormonal method as assessed
spermatogenesis and increase the rate of by opinion polls is high.
azoospermia, testosterone is combined with  After academic research established the principle
either progestins or GnRH antagonists. of hormonal male contraception, it remains to be
 All effective approaches tested so far require seen whether the pharmaceutical industry will
injections or implantations. Self-administered enter the field of male contraception again.

22.10 References Intramuscular testosterone


enanthate plus very low dosage oral
administration of levonorgestrel
and testosterone induces more rapid
Alvarez-Sanchez F, Faundes A, Brache levonorgestrel suppresses and effective suppression of
V, Leon P (1977) Attainment and spermatogenesis without causing spermatogenesis than testosterone
maintenance of azoospermia with weight gain in normal young men: a alone: a promising male
combined monthly injections of randomized clinical trial. J Androl contraceptive approach. J Clin
depot medroxyprogesterone acetate 26:405413 Endocr Metab 81:757762
and testosterone enanthate.
Contraception 15:635648 Anderson RA, Baird DT (1997) Behre HM, Nashan D, Hubert W,
Progress towards a male pill. IPPF Nieschlag E (1992) Depot
Amory JK, Leonard TW, Page ST, Med Bull 31:15 gonadotropin-releasing hormone
OToole E, McKenna MJ, Bremner agonist blunts the androgen-
WJ (2009) Oral administration of Anderson RA, Van Der Spuy ZM,
induced suppression of
the GnRH antagonist acyline, in a Dada OA, Tregoning SK, Zinn PM,
spermatogenesis in a clinical trial of
GIPET-enhanced tablet form, Adeniji OA, Fakoya TA, Smith KB,
male contraception. J Clin Endocr
acutely suppresses serum Baird DT (2002a) Investigation of
Metab 74:8490
testosterone in normal men: single- hormonal male contraception in
African men: suppression of Behre HM, Bckers A, Schlingheider A,
dose pharmacokinetics and
spermatogenesis by oral desogestrel Nieschlag E (1994) Sustained
pharmacodynamics. Cancer
with depot testosterone. Hum suppression of serum LH, FSH and
Chemother Pharmacol 64:641645
Reprod 17:28692877 testosterone and increase of high-
Anawalt BD, Bebb RA, Bremner WJ, density lipoprotein cholesterol by
Matsumoto AM (1999) A lower Anderson RA, Kinniburgh D,
daily injections of the GnRH
dosage levonorgestrel and Baird DT (2002b) Suppression of
antagonist Cetrorelix over 8 days in
testosterone combination effectively spermatogenesis by etonogestrel
normal men. Clin Endocr
implants with depot testosterone:
suppresses spermatogenesis and 40:241248
circulating gonadotrophin levels potential for long-acting male
contraception. J Clin Endocr Metab Behre HM, Baus S, Kliesch S, Keck C,
with fewer metabolic effects than
87:36403649 Simoni M, Nieschlag E (1995)
higher dosage combinations.
Potential of testosterone buciclate
J Androl 20:407414 Aribarg A, Sukcharoen N, Chanprasit for male contraception: endocrine
Anawalt BD, Herbst KL, Matsumoto Y, Ngeamvijawat J, Kriangsinyos R differences between responders and
AM, Mulders TM, Coelingh- (1996) Suppression of nonresponders. J Clin Endocr Metab
Bennink HJ, Bremner WJ (2000) spermatogenesis by testosterone 80:23942403
Desogestrel plus testosterone enanthate in Thai men. J Med Assoc
Thai 79:624629 Behre HM, Kliesch S, Lemcke B,
effectively suppresses Nieschlag E (2001) Suppression of
spermatogenesis but also causes Arysad, KM (1993) Sperm function in spermatogenesis to azoospermia
modest weight gain and high- Indonesian men treated with by combined administration
density lipoprotein suppression. testosterone enanthate. Int J Androl of GnRH antagonist and
Fertil Steril 74:707714 16:355361 19-nortestosterone cannot be
Anawalt BD, Amory JK, Herbst KL, Bebb RA, Anawalt BD, Christensen RB, maintained by 19-nortestosterone
Coviello AD, Page ST, Bremner WJ, Paulsen CA, Bremner WJ, alone in normal men. Hum Reprod
Matsumoto AM (2005) Matsumoto AM (1996) Combined 16:25702577

488
Chapter 22: Role of testosterone in male contraception

Bhasin S, Berman N, Swerdloff RS ethinyltestosterone and 19-nor-17a- L, Ho PC, Anderson RA (2000)


(1994) Follicle-stimulating methyltestosterone. J Am Chem Soc Would women trust their partners
hormone (FSH) escape during 76:40924094 to use a male pill? Hum Reprod
chronic gonadotropin-releasing 15:646649
Ersheng G, Cuihong L, Youlun G,
hormone (GnRH) agonist and
Lamei L, Changhai H (1999) Grimes DA, Lopez LM, Gallo MF,
testosterone treatment. J Androl
Inhibiting effects of sino-implant Halpern V, Nanda K, Schulz KF
15:386391
plus testosterone undecanoate on (2007) Steroid hormones for
Boccon-Gibod L, van der Meulen E, spermatogenesis in Chinese men. contraception in men. Cochrane
Persson BE (2011) An update on the Reprod Contracept 10:98105 Database Syst Rev 18:CD004316
use of gonadotropin-releasing
Faundes A, Brache V, Leon P, Schmidt Gu Y, Liang X, Wu W, Liu M, Song S,
hormone antagonists in prostate
F, Alvarez-Sanchez F (1981) Sperm Cheng L, Bo L, Xiong C, Wang X,
cancer. Ther Adv Urol 3:127140
suppression with monthly injections Liu X, Peng L, Yao K (2009)
Brady BM, Walton M, Hollow N, of medroxyprogesterone acetate Multicenter contraceptive efficacy
Kicman AT, Baird DT, Anderson RA combined with testosterone trial of injectable testosterone
(2004) Depot testosterone with enanthate at a high dose (500 mg). undecanoate in Chinese men. J Clin
etonogestrel implants result in Int J Androl 4:235245 Endocrinol Metab 94:19101915
induction of azoospermia in all men
for long-term contraception. Hum Fogh M, Corker CS, Hunter WM, Gu YQ, Wang X-H, Xu D, Peng L,
Reprod 19:26582667 McLean H, Philip J, Schon G, Cheng L-F, Huang M-K, Huang Z-J,
Skakkebaek NE (1979) The effects Zhang G-Y (2003) A multicenter
Brenner PF, Mishell DR Jr, of low doses of cyproterone acetate contraceptive efficacy study of
Bernstein GS, Ortiz A (1977) Study on some functions of the injectable testosterone undecanoate
of medroxyprogesterone acetate and reproductive system in normal men. in healthy Chinese men. J Clin
testosterone enanthate as a male Acta Endocrinol 91:545 Endocr Metab 88:562568
contraceptive. Contraception
15:679691 Fogh M, Corker CS, McLean H (1980) Gu YQ, Tong JS, Ma DZ, Wang XH,
Clinical trial with levonorgestrel Yuan D, Tang WH, Bremner WJ
Bchter D, von Eckardstein S, von and testosterone enanthate for male (2004) Male hormonal
Eckardstein A, Kamischke A, fertility control. Acta Endocrinol contraception: effects of injections
Simoni M, Behre HM, Nieschlag E 95:251257 of testosterone undecanoate and
(1999) Clinical trial of transdermal depot medroxyprogesterone acetate
testosterone and oral levonorgestrel Frick J, Bartsch G, Weiske WH (1977)
at eight-week intervals in Chinese
for male contraception. J Clin The effect of monthly depot
men. J Clin Endocrinol Metab
Endocr Metab 84:12441249 medroxyprogesterone acetate and
89:22542262
testosterone on human
Cao J, Yuan J, Jin W (1996) Clinical
spermatogenesis. II. High initial Guerin JF, Rollet J (1988) Inhibition of
trial of an anti-fertility method with
dose. Contraception 15:669677 spermatogenesis in men using
testosterone enanthate in normal
various combinations of oral
men. Chung Hua I Hsueh Tsa Chih Frick J, Danner C, Kunit G, Joos H,
progestagens and percutaneous
76:335337 Kohle R (1982) Spermatogenesis in
or oral androgens. Int J Androl
men treated with injections of
CONRAD (2011) Male Hormonal 11:187199
medroxyprogesterone acetate
Contraceptive Trial Ending Early: Gui YL, He CH, Amory JK, Bremner
combined with testosterone
Innovative Study is a Collaboration
enanthate. Int J Androl 5:246252 WJ, Zheng EX, Yang J, Yang PJ,
of WHO and CONRAD, April 22, Gao ES (2004) Male hormonal
2011. Available at: http://www. Gaw Gonzalo IT, Swerdloff RS, Nelson AL,
contraception: suppression of
conrad.org/news-pressreleases-63. Clevenger B, Garcia R, Berman N,
spermatogenesis by injectable
html (accessed Jan 20, 2012) Wang C (2002) Levonorgestrel
testosterone undecanoate alone or
implants (Norplant II) for male
Cooper TG, Yeung CH (2010) with levonorgestrel implants in
contraception clinical trials:
Pharmacological approaches to Chinese men. J Androl 25:720727
combination with transdermal and
male contraception. In: Nieschlag E, Hair WM, Kitteridge K, Wu FCW
injectable testosterone. J Clin Endocr
Behre HM, Nieschlag S (eds)
Metab 87:35623572 (1999) A new male contraceptive
Andrology. Male Reproductive pill/patch combination oral
Health and Dysfunction, 3rd edn. Glasier A (2010) Acceptability of desogestrel and transdermal
Springer-Verlag, Berlin, pp 589599 contraception for men: a review. testosterone: suppression of
Djerassi C, Miramontes L, Rosenkranz Contraception 82:453450 gonadotropins and spermatogenesis
G, Sonheimer F (1954) Steroids Glasier AF, Anakwe R, Everington D, in men. In: The Endocrine
LIV. Synthesis of 19-nor-17a- Martin CW, van der Spuy Z, Cheng Societys 81st Annual Meeting; 1999

489
Chapter 22: Role of testosterone in male contraception

June 1215; San Diego, CA, USA; norethisterone preparations. J Clin McLachlan RI, ODonnell L, Stanton
Poster P3374 Endocr Metab 87:530539 PG, Balourdos G, Frydenberg M,
Handelsman DJ, Conway AJ, Boylan LM Kinniburgh D, Anderson RA, Baird DeKretser DM, Robertson DM
(1992) Suppression of human DT (2001) Suppression of (2002) Effects of testosterone
spermatogenesis by testosterone spermatogenesis with desogestrel enanthate plus
implants. J Clin Endocr Metab and testosterone pellets is not medroxyprogesterone acetate on
75:13261332 enhanced by addition of finasteride. semen quality, reproductive
J Androl 22:8895 hormones and germ cell
Handelsman DJ, Conway AJ, Howe CJ, populations in normal young men.
Turner L, Mackey MA (1996) Kinniburgh D, Zhu H, Cheng L, J Clin Endocrinol Metab 87:546556
Establishing the minimum Kicman AT, Baird DT, Anderson RA
effective dose and additive Melo JF, Coutinho EM (1977)
(2002) Oral desogestrel with
effects of depot progestin in Inhibition of spermatogenesis in
testosterone pellets induces
suppression of human men with monthly injections of
consistent suppression of
spermatogenesis by a testosterone medroxyprogesterone acetate and
spermatogenesis to azoospermia in
depot. J Clin Endocrinol Metab testosterone enanthate.
both Caucasian and Chinese men.
81:41134121 Contraception 15:627634
Hum Reprod 17:14901501
Meriggiola MC, Bremner WJ, Paulsen
Heinemann K, Saad F, Wiesemes M, Knuth UA, Behre, HM, Belkien L, CA, Valdiserri A, Incorvala L,
White S, Heinemann L (2005) Bents H, Nieschlag E (1985) Clinical Motta R, Pavani A, Capelli M,
Attitudes toward male fertility trial of 19-nortestosterone- Flamigni C (1996) A combined
control: results of a multinational hexoxyphenylpropionate (Anadur) regimen of cyproterone acetate and
survey on four continents. Hum for male fertility regulation. Fertil testosterone enanthate as a
Reprod 20:549556 Steril 44:814821 potentially highly effective male
Kamischke A, Nieschlag E (2004) Knuth UA, Yeung CH, Nieschlag E (1989) contraceptive. J Clin Endocr Metab
Progress towards hormonal male Combination of 19-nortestosterone- 81:30183023
contraception.Trends Pharmacol Sci hexyoxyphenylpropionate (Anadur) Meriggiola MC, Bremner WJ,
25:4957 and depot-medroxyprogesterone- Costantino A, Pavani A, Capelli M,
Kamischke A, Plger D, Venherm S, acetate (Clinovir) for male Flamigni C (1997) An oral regimen
von Eckardstein S, von Eckardstein A, contraception. Fertil Steril of cyproterone acetate and
Nieschlag E (2000a) Intramuscular 51:10111018 testosterone undecanoate for
testosterone undecanoate with or Liu PY, Swerdloff RS, Christenson PD, spermatogenic suppression in men.
without oral levonorgestrel: a Handelsman DJ, Wang C; Fertil Steril 68:844850
randomized placebo-controlled Hormonal Male Contraception Meriggiola MC, Bremner WJ,
feasibility study for male Summit Group (2006) Rate, extent Costantino A, Di Cintio G,
contraception. Clin Endocrinol and modifiers of spermatogenic Flamigni C (1998) Low dose of
53:4352 recovery after hormonal male cyproterone acetate and
Kamischke A, Diebcker J, Nieschlag E contraception: an integrated testosterone enanthate for
(2000b) Potential of norethisterone analysis. Lancet 367:14121420 contraception in men. Hum Reprod
enanthate for male contraception:
Martin CW, Anderson RA, Cheng L, 13:12251229
pharmacokinetics and suppression
Ho PC, van der Spuy Z, Smith KB, Meriggiola MC, Costantino A,
of pituitary and gonadal function.
Glasier AF, Everington D, Baird DT Bremner WJ, Morselli-Labate AM
Clin Endocrinol 53:351358
(2000) Potential impact of (2002a) Higher testosterone dose
Kamischke A, Venherm S, Plger D, hormonal male contraception: impairs sperm suppression induced
von Eckardstein S, Nieschlag E cross-cultural implications for by a combined androgen-progestin
(2001) Intramuscular testosterone development of novel preparations. regimen. J Androl 23:684690
undecanoate and norethisterone Hum Reprod 15:637645 Meriggiola MC, Bremner WJ,
enanthate in a clinical trial for male
McLachlan RI, McDonald J, Costantino A, Bertaccini A,
contraception. J Clin Endocr Metab
Rushford D, Robertson DM, Morselli-Labate AM, Huebler D,
86:303309
Garrett C, Baker HW (2000) Kaufmann G, Oettel M, Flamigni C
Kamischke A, Heuermann T, Krger K, Efficacy and acceptability of (2002b) Twenty-one day
von Eckardstein S, Schellschmidt I, testosterone implants, alone or in administration of dienogest
Rbig A, Nieschlag E (2002) An combination with a 5a-reductase reversibly suppresses gonadotropins
effective hormonal male inhibitor, for male hormonal and testosterone in normal men.
contraceptive using testosterone contraception. Contraception J Clin Endocrinol Metab
undecanoate with oral or injectable 62:7378 87:21072113

490
Chapter 22: Role of testosterone in male contraception

Meriggiola MC, Costantino A, Variability in sperm suppression Nieschlag E, Vorona E, Wenk M,


Cerpolini S, Bremner WJ, Huebler D, during testosterone administration Hemker AK, Kamischke A,
Morselli-Labate AM, Kirsch B, to adult monkeys is related to Zitzmann M (2011) Hormonal
Bertaccini A, Pelusi C, Pelusi G follicle stimulating hormone male contraception in men
(2003) Testosterone undecanoate suppression and not to with normal and subnormal
maintains spermatogenic intratesticular androgens. J Clin semen parameters. Int J Androl
suppression induced by cyproterone Endocrinol Metab 87:33993406 34:556567
acetate plus testosterone undecanoate Nieschlag E (1986) Reasons for Page ST, Amory JK, Anawalt BD, Irwig
in normal men. J Clin Endocrinol abandoning immunization against MS, Brockenbrough AT,
Metab 88:58185826 FSH as an approach to male fertility Matsumoto AM, Bremner WJ
Meriggiola MC, Costantino A, Saad F, regulation. In: Zatuchni GI, (2006) Testosterone gel combined
DEmidio L, Morselli Labate AM, Goldsmith A, Spieler JM, Sciarra J with depomedroxyprogesterone
Bertaccini A, Bremner WJ, Rudolph I, (eds) Male Contraception: acetate is an effective male
Ernst M, Kirsch B, Martorana G, Advances and Future Prospects. hormonal contraceptive regimen
Pelusi G (2005) Norethisterone Harper & Row, Philadelphia, and is not enhanced by the
enanthate plus testosterone pp 395400 addition of a GnRH antagonist.
undecanoate for male J Clin Endocrinol Metab
Nieschlag E (2009) Male hormonal
contraception: effects of various 91:43744380
contraception: loves labours lost?
injection intervals on Pangkahila W (1991) Reversible
(Editorial) J Clin Endocrinol Metab
spermatogenesis, reproductive
94:18901892 azoospermia induced by an
hormones, testis, and prostate. androgen-progestin combination
J Clin Endocrinol Metab Nieschlag E, Behre HM (1996)
regimen in Indonesian men. Int
90:20052014 Hormonal male contraception:
J Androl 14:248256
suppression of spermatogenesis
Misro MM, Chaki SP, Kaushik MC, Patanelli DJ (ed) (1978) Hormonal
with GnRH antagonists and
Nandan D (2009) Trials for Control of Male Fertility (Workshop
testosterone. In: Filicori M,
development of once-a-month Proceedings), NIH Publication No.
Flamigni C (eds) Treatment with
injectable, hormonal male 781097. US Department of Health,
GnRH Analogs: Controversies and
contraceptive using dienogest plus Education, and Welfare (NIH),
Perspectives. Parthenon, London,
testosterone undecanoate: dose Bethesda, MD
pp 243248
standardization, efficacy and
reversibility studies in rats. Nieschlag E, Hoogen H, Blk M, Pavlou SN, Wakefield G, Schlechter NL,
Contraception 79:488497 Schuster H, Wickings EJ (1978) Lindner J, Souza KH, Kamilaris TC,
Clinical trial with testosterone Konidaris S, Rivier JE, Vale WW,
Moltz L, Rmmler A, Post K, Toglia M (1989) Mode of
undecanoate for male fertility
Schwartz U, Hammerstein J (1980) suppression of pituitary and
control. Contraception 18:607614
Medium dose cyproterone acetate gonadal function after acute or
(CPA): effects on hormone Nieschlag E, Behre HM, Weinbauer GF prolonged administration of a
secretion and on spermatogenesis in (1992) Hormonal male luteinizing hormone-releasing
man. Contraception 21:393 contraception: a real chance? In: hormone antagonist in normal men.
Nieschlag E, Habenicht UF (eds) J Clin Endocrinol Metab
Mommers E, Kersemaeker WM,
Spermatogenesis Fertilization 73:13601369
Elliesen J, Kepers M, Apter D, Behre
Contraception. Molecular, Cellular
HM, Beynon J, Bouloux PM, Pavlou SN, Herodotou D, Curtain M,
and Endocrine Events in Male
Costantino A, Gerbershagen HP, Minaretzis D (1994) Complete
Reproduction. Springer, Heidelberg,
Gronlund L, Heger-Mahn D, suppression of spermatogenesis by
pp 477501
Huhtaniemi I, Koldewihn EL, Lange co-administration of a GnRH
C, Lindenberg S, Meriggiola MC, Nieschlag E; 10th Summit Meeting antagonist plus a physiologic dose
Meuleman E, Mulder PFA, Group (2007) 10th Summit Meeting of testosterone. In: Proceedings of
Nieschlag E, Perheentupa A, consensus: recommendations for the 76th Meeting of the Endocrine
Solomon A, Visl L, Wu FC, regulatory approval for hormonal Society, Anaheim, CA, p 1324
Zitzmann M (2008) Male hormonal male contraception. October 2223, Pllnen P, Nikkanen V, Huhtaniemi I
contraception: a double-blind, 2006. Contraception 75:166167 (2001) Combination of
placebo-controlled study. J Clin Nieschlag E, Nieschag S, Behre HM subcutaneous levonorgestrel
Endocrinol Metab 93:25722580 (eds) (2010) Andrology. Male implants and transdermal
Narula A, Gu Y-Q, ODonnell L, Reproductive Health and dihydrotestosterone gel for male
Stanton PG, Robertson DM, Dysfunction, 3rd edn. Springer- hormonal contraception. Int
McLachlan RI, Bremner WJ (2002) Verlag, Berlin J Androl 24:369380

491
Chapter 22: Role of testosterone in male contraception

Qoubaitary A, Meriggiola C, Ng CM, Contraceptive efficacy of a depot apoptosis. J Clin Endocrinol Metab
Lumbreras L, Cerpolini S, Pelusi G, progestin and androgen 92:32923304
Christensen PD, Hull L, combination in men. J Clin Endocr
Weinbauer GF, Nieschlag E (1996) The
Swerdloff RS, Wang C (2006) Metab 88:46594667
Leydig cell as a target for male
Pharmacokinetics of testosterone von Eckardstein S, Schmidt A, contraception. In: Payne AH, Hardy
undecanoate injected alone or in Kamischke A, Simoni M, Gromoll J, MP, Russell LD (eds) The Leydig
combination with norethisterone Nieschlag E (2002) CAG repeat Cell. Cache River Press, Vienna, IL,
enanthate in healthy men. J Androl length in the androgen receptor pp 629662
27:853867 gene and gonadotropin suppression
Weinbauer GF, Surmann FJ, Nieschlag
Reddy PR, Rao JM (1972) Reversible influence the effectiveness of
E (1987) Suppression of
antifertility action of testosterone hormonal male contraception. Clin
spermatogenesis in a nonhuman
propionate in human males. Endocrinol 57:647655
primate (Macaca fascicularis) by
Contraception 5:295301 von Eckardstein S, No G, Brache V, concomitant gonadotropin-
Schearer SB, Alvarez-Sanches F, Nieschlag E, Croxatto H, Alvarez F, releasing hormone (GnRH)
Anselmo G, Brenner P, Coutinho E, Moo-Young A, Sivin I, Kumar N, antagonist and testosterone
Lathen-Faundes A, Frick J, Heinild Small M, Sundaram K (2003) treatment. Acta Endocrinol
B, Johansson EDB (1978) Hormonal A clinical trial of 7a-methyl-19- 114:138146
contraception for men. Int J Androl nortestosterone implants for
2 (Suppl 2):680712 possible use as a long acting Weinbauer GF, Khurshid S,
contraceptive for men. J Clin Fingscheidt U, Nieschlag E (1989)
Schrmeyer T, Knuth UA, Belkien L, Sustained inhibition of sperm
Nieschlag E (1984) Reversible Endocrinol Metab 88:52325239
production and inhibin secretion
azoospermia induced by the Waites GMH (2003) Development of induced by a gonadotropin-
anabolic steroid 19-nortestosterone. methods of male contraception: the releasing hormone antagonist and
Lancet 25:417420 impact of the World Health delayed testosterone substitution in
Singh S, Sedh G, Hussain R (2010) Organization Task Force. Fertil non-human primates (Macaca
Unintended pregnancy: worldwide Steril 80:115 fascicularis). J Endocrinol
levels, trends, and outcomes. Stud Walton MJ, Kumar N, Baird DT, 123:303310
Fam Plann 41:241250 Ludlow H, Anderson RA (2007) Weinbauer GF, Limberger A, Behre
Soufir JC, Meduri G, Ziyyat A (2011) 7alpha-methyl-19-nortestosterone HM, Nieschlag E (1994) Can
Spermatogenetic inhibition in men (MENT) vs testosterone in testosterone alone maintain the
taking a combination of oral combination with etonogestrel gonadotropin-releasing hormone
medroxyprogesterone acetate and implants for spermatogenic antagonist-induced suppression
percutaneous testosterone as a male suppression in healthy men. of spermatogenesis in the non-
contraceptive method. Hum Reprod J Androl 28:679688 human primate? J Endocrinol
26:17081714 Wang C, Yeung KK (1980) Use of low- 142:485495
Srinath BR, Wickings EJ, Witting CH, dosage oral cyproterone acetate as a Weinbauer GF, Schlatt S, Walter V,
Nieschlag E (1983) Active male contraceptive. Contraception Nieschlag E (2001) Testosterone-
immunization with follicle 21:245 induced inhibition of
stimulating hormone for fertility Wang C, Wang XH, Nelson AL (2006) spermatogenesis is more closely
control: a 4 1/2-year study in male Levonorgestrel implants enhanced related to suppression of FSH than
rhesus monkeys. Fertil Steril the suppression of spermatogenesis to testicular androgen levels in the
40:110117 by testosterone implants: cynomolgus monkey model
Swerdloff RS, Bagatell CJ, Wang C, comparison between Chinese and (Macaca fascicularis). J Endocrinol
Anawalt BD, Berman N, Steiner B, non-Chinese men. J Clin Endocrinol 168:2538
Bremner WJ (1998) Suppression of Metab 91:460470 Weston GC, Schlipalius ML,
spermatogenesis in man induced by Wang C, Cui YG, Wang XH, Jia Y, Bhuinneain MN, Vollenhoven BJ
Nal-Glu gonadotropin releasing Sinha Hikim A, Lue YH, Tong JS, (2002) Will Australian men use
hormone antagonist and Qian LX, Sha JH, Zhou ZM, Hull L, male hormonal contraception?
testosterone enanthate (TE) is Leung A, Swerdloff RS (2007) A survey of a postpartum
maintained by TE alone. J Clin Transient scrotal hyperthermia and population. Med J Aust 176:204205
Endocr Metab 83:35273533 levonorgestrel enhance WHO (19721995) Special Programme
Turner L, Wishart S, Conway AJ, testosterone-induced of Research, Development and
Liu PY, Forbes E, McLachlan RI, spermatogenesis suppression in Research Training in Human
Handelsman DJ (2003) men through increased germ cell Reproduction, Annual and Biannual

492
Chapter 22: Role of testosterone in male contraception

Reports. World Health WHO Task Force on Methods for the metabolism. J Clin Endocr Metab
Organization, Geneva Regulation of Male Fertility (1996) 84:12122
WHO Task Force on Methods for the Contraceptive efficacy of Zhang GY, Gu YQ, Wang XH, Cui YG,
Regulation of Male Fertility (1990) testosterone-induced azoospermia Bremner WJ (1999) A clinical trial
Contraceptive efficacy of and oligozoospermia in normal of injectable testosterone
testosterone-induced azoospermia men. Fertil Steril 65:821829 undecanoate as a potential male
in normal men. Lancet 336:955959 Wu FC, Aitken RJ (1989) Suppression contraceptive in normal Chinese
WHO Task Force on Methods for the of sperm function by depot men. J Clin Endocrinol Metab
Regulation of Male Fertility (1993) medroxyprogesterone acetate and 84:36423647
Comparison of two androgens plus testosterone enanthate in steroid Zhang L, Shal IH, Liu Y, Vogelsong
depot-medroxyprogesterone acetate male contraception. Fertil Steril KM, Zhang L (2006) The
for suppression to azoospermia in 51:691698 acceptability of an injectable,
Indonesian men. Fertil Steril Wu FCW, Balasubramanian R, once-a-month male
60:10621068 Mulders TMT, Coelingh-Bennink contraceptive in China.
WHO Task Force on Methods for the HJT (1999) Oral progestogen Contraception 73:548553
Regulation of Male Fertility (1995) combined with testosterone as a Zitzmann M, Junker R, Kamischke A,
Rates of testosterone-induced potential male contraceptive: Nieschlag E (2002) Contraceptive
suppression to severe additive effects between desogestrel steroids influence the
oligozoospermia or azoospermia in and testosterone enanthate in hemostatic activation state
two multinational clinical studies. suppression of spermatogenesis, in healthy men. J Androl
Int J Androl 18:157165 pituitary-testicular axis, and lipid 23:503511

493
Chapter
Testosterone use in women

23 Susan R. Davis

23.1 Introduction 494 23.4.4 Effects of testosterone therapy on mood


23.2 Physiology 495 and well-being 504
23.2.1 Testosterone physiology in the female 23.4.5 Effects of testosterone therapy on
reproductive years 495 cardiovascular function and
23.2.2 Physiological and non-physiological cardiovascular disease risk 504
causes of lower testosterone levels 23.4.6 Effects on bone and lean mass 504
in women 496 23.4.7 Effects of testosterone on
23.3 Clinical associations with endogenous cognition 504
testosterone levels in women: findings 23.4.8 Use of testosterone for treatment
from observational studies 498 of vulval lichen sclerosis 505
23.3.1 Testosterone and sexual function 498 23.5 Safety of testosterone therapy for
23.3.2 Testosterone and well-being 498 women 505
23.3.3 Testosterone and cardiovascular 23.5.1 Testosterone and adverse androgenic
disease 499 effects 505
23.3.4 Testosterone and osteoporosis 499 23.5.2 Testosterone and breast cancer risk 505
23.3.5 Testosterone and cognitive 23.5.3 Testosterone and endometrial cancer
function 500 risk 506
23.4 Effects of testosterone: findings from 23.5.4 Other safety concerns 507
randomized, controlled trials 500 23.6 Appropriate candidates for testosterone
23.4.1 The use of testosterone for the therapy 507
treatment of female sexual 23.6.1 Identifying women likely to benefit from
dysfunction 500 testosterone therapy 507
23.4.2 The need to treat hypoactive sexual 23.6.2 Assessment of women presenting with
desire disorder 500 low sexual desire 508
23.4.3 Efficacy of testosterone for treatment 23.7 Treatment options 509
of hypoactive sexual desire disorder: 23.8 Conclusions 509
findings from randomized, controlled 23.9 Key messages 510
trials 501 23.10 References 510

23.1 Introduction the mid-reproductive years, and are low in women


The role of testosterone in women and its potential as who have experienced surgical menopause, premature
a therapeutic agent continues to attract controversy. ovarian failure and hypopituitarism. The primary
Testosterone levels in women decline with age from outcome of clinical trials on the effects of testosterone

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

494
Chapter 23: Testosterone use in women

in women has been on treatment of female sexual regarding elective bilateral oophorectomy in peri/
dysfunction; notably that characterized by loss of postmenopausal women undergoing hysterectomy.
sexual desire. There is substantial evidence that judi- Testosterone can undergo 5a-reduction to form
cious testosterone therapy is effective for the treat- DHT. Dihydrotestosterone can then be converted to
ment of loss of sexual desire in postmenopausal androstane-3a,17b-diol (3a-diol), which in turn can be
women and women approaching the late reproductive glucuronidated at carbons 3 or 17 to form 3a-diol-
years, as well as women with ovarian or pituitary 3-glucuronide (3a-diol,3G) or 3a-diol-17-glucuronide
failure, with doses given that achieve circulating levels (3a-diol,17G), of which 3a-diol,17G is the predominant
close to the physiological range for young reproductive form. Plasma 3a-diol,17G is a marker of peripheral
women. The greatest body of clinical data is from androgen action and is significantly elevated in idio-
studies of transdermal testosterone in postmenopausal pathic hirsutism (Horton et al. 1982). Alternatively,
women. The link between postmenopausal estrogen- testosterone can be aromatized to estradiol in extra-
progestin use and both breast cancer and cardiovascu- gonadal tissues including the brain, adipose tissue, vas-
lar disease has created a level of concern regarding any cular endothelium and bone (Simpson and Davis 2001).
form of hormone use in women. Objective data show Therefore, maintenance of physiological circulating tes-
no adverse cardiovascular effects of transdermal testos- tosterone levels in women ensures adequate supply of
terone therapy, and cumulative data from RCTs do not substrate for estrogen biosynthesis in extra-gonadal
indicate increased cancer risk in women treated with sites, such as bone, in which high tissue estrogen concen-
testosterone. A recent Cochrane Review (Somboon- trations may be physiologically important. That the
porn et al. 2005) has thoroughly evaluated the evidence primary source of estrogen in extra-gonadal sites is
for the effects of testosterone added to estrogen therapy that produced locally from androgens also explains the
in postmenopausal women. This chapter reviews the apparent threshold dose of estrogen replacement after
physiology of testosterone in women, summarizes the menopause below which bone loss continues, and the
findings from observational studies and clinical trials, observation that, whereas standard estrogen replace-
and considers indications for use. ment therapy has little effect on libido (Utian 1972;
Campbell and Whitehead 1977), most parameters of
sexuality improve when extremely high doses of estro-
23.2 Physiology gen are administered (Davis et al. 1995).
23.2.1 Testosterone physiology in the There is significant cyclicity in plasma levels of
androstenedione and testosterone in regularly ovulat-
female reproductive years ing women, with increases in the mean circulating
In healthy, young, reproductive-aged women testoster- levels of both of these hormones in the middle third
one circulates in nanomolar concentrations, in contrast of the menstrual cycle (Mushayandebvu et al. 1996).
to picomolar levels of estradiol. The production of tes- This is followed by a second rise in androstenedione
tosterone in premenopausal women is of the order of 0.2 production by the corpus luteum during the late
to 0.25 mg per day (Longcope 1986). Approximately half luteal phase. Ovarian androgens are produced by the
of the circulating testosterone is produced by the ovaries, thecal cells under the control of LH.
with the remainder produced by peripheral conversion Under normal physiological conditions, only 12%
of pre-androgens to testosterone, with androstenedione of total circulating testosterone in women circulates
being the main precursor. Whether there is direct secre- unbound to plasma proteins. The rest is bound by sex
tion of testosterone by the adrenals continues to be hormone-binding globulin (SHBG) and albumin, with
debated. The pre-androgens, androstenedione and SHBG binding 66% of total circulating testosterone in
DHEA, are produced by both the ovaries and the adre- healthy women (Dunn et al. 1981). The binding affinity
nals, with the adrenals also being the main site of pro- for steroids bound by SHBG is DHT > testosterone
duction of DHEAS. Circulating DHEAS is an important > androstenediol > estradiol > estrone (Dunn et al.
precursor for the ovarian intrafollicular production 1981). SHBG also weakly binds DHEA, but not DHEAS
of testosterone and DHT (Haning et al. 1993). (Dunn et al. 1981). Therefore, variations in the plasma
Whether the postmenopausal ovary continues to levels of SHBG will influence the amount of unbound
secrete testosterone remains contentious, as dis- testosterone. It is frequently stated that endogenous
cussed below. Hence, there is ongoing debate testosterone lowers, and estrogen increases, SHBG

495
Chapter 23: Testosterone use in women

production. However, in-vitro studies show that DHT and the ratio of total testosterone to SHBG do
extremely high levels of estradiol and testosterone are not change (Burger et al. 1995; Davison et al. 2005a).
required to influence SHBG biosynthesis and release Most studies indicate that the postmenopausal
from the hepatocytes (Edmunds et al. 1990; Loukovaara ovary in many women continues to produce testoster-
et al. 1995). Taken together, in-vitro studies indicate no one. Acutely following oophorectomy there is a decline
effect of estradiol and testosterone on hepatocyte SHBG in testosterone, with both testosterone and androstene-
production at physiological concentrations seen in dione decreasing acutely by about 50% (Judd et al.
women; high hepatic concentrations of estradiol, as seen 1994), and postmenopausal women who have under-
with oral estrogen, might increase SHBG biosynthesis, gone bilateral oophorectomy have been found to have
but there are conflicting findings for effects of supra- lower total and free testosterone levels than women
physiological testosterone on hepatocyte SHBG produc- who have not (Laughlin et al. 2000; Davison et al.
tion (Loukovaara et al. 1995). There is also a widely held 2005a). Peripheral and ovarian vein sampling of
belief that insulin suppresses SHBG production (Pugeat women undergoing bilateral oophorectomy with and
et al. 2000). However, insulin does not directly regulate without GnRH therapy has demonstrated that the post-
SHBG gene expression in vitro, but rather fructose and menopausal ovary is hormonally active and contributes
glucose, as well as peroxisome-proliferator receptor significantly to the pool of testosterone in women (Judd
gamma are key regulators of human SHBG biosynthesis et al. 1974; Sluijmer et al. 1995; Fogle et al. 2007). In
(Selva et al. 2007; Selva and Hammond 2009). Fructose contrast, other investigators believe that the postmeno-
and glucose-induced lipogenesis reduces hepatocyte pausal ovary does not produce testosterone (Couzinet
nuclear factor 4alpha levels, which in turn attenuate et al. 2001; Labrie et al. 2010). In a study comparing
SHBG expression (Selva et al. 2007). This in part healthy postmenopausal women with and without
explains why SHBG is a sensitive biomarker of the oophorectomy and/or adrenalectomy, adrenalectomy
metabolic disturbances associated with increased fruc- was associated with total and bioavailable testosterone
tose consumption. below the detection limit; whereas androstenedione was
reduced but measurable, even in women who had
undergone both oophorectomy and adrenalectomy
23.2.2 Physiological and non-physiological (Couzinet et al. 2001). In this study the number of
women providing data was small, and there was marked
causes of lower testosterone levels in women variability in testosterone levels. Another more recent
The main cause of lowered testosterone levels in women study of oophorectomized and ovary-intact women has
is the natural decline with age. The mean circulating shown wide ranges of testosterone levels in these popu-
levels of total and free testosterone decline continuously lations, and although an argument was mounted that
with increasing age from the early reproductive years, the study demonstrated no direct contribution of tes-
such that the levels of women in their forties are approxi- tosterone from the postmenopausal ovary, this was not
mately half of those of women in their twenties, as shown supported by the study data (Labrie et al. 2010).
in Fig. 23.1 (Zumoff et al. 1995; Davison et al. 2005a). Hysterectomy has also been associated with lower
Androstenedione and DHEAS levels also fall linearly circulating testosterone in women (Laughlin et al.
with age (Davison et al. 2005a), and this may contribute 2000; Davison et al. 2005a). Other iatrogenic causes
to the decline in the level of their main metabolite, of low testosterone include chemical oophorectomy;
testosterone. In the late reproductive years there is for example, the use of GnRH antagonists for the
failure of the mid-cycle rise in free testosterone that treatment of fibroids or endometriosis, postche-
characterizes the menstrual cycle in young ovulating motherapy or radiotherapy, and the administration
women (Mushayandebvu et al. 1996). This occurs des- of oral estrogens or oral glucocorticosteroids. In gen-
pite preservation of normal free testosterone levels at eral, circulating free testosterone is suppressed in
other phases of the cycle. The mean plasma concentra- women using either the combined oral contraceptive
tions of testosterone in women transiting the menopause pill or oral estrogen as hormone replacement therapy
are also significantly lower than younger ovulating (Mathur et al. 1985; Krug et al. 1994). This is a result
women sampled in the early follicular phase. Testoster- of an increase in SHBG combined with suppression of
one levels do not fall at menopause. Across the perimeno- LH production by the pituitary and, hence, decreased
pausal period total testosterone, androstenedione, stimulus for the ovarian stromal production of

496
4 4

Total testosterone nmol/l

Total testosterone nmol/l


Total testosterone 3 3

2 2

1 1

0 0
20 30 40 50 60 70 80 1824 2534 3544 4554 5564 6575
Age, years Age group, years
15 15

DHEAS
DHEAS mol/l

10 10

DHEAS mol/l
5 5

0 0
20 30 40 50 60 70 80 1824 2534 3544 4554 5564 6575
Age, years Age group, years
60 60
Free testosterone
Free testosterone pmol/I

Free testosterone pmol/I

40 40

20 20

0 0
20 30 40 50 60 70 80 1824 2534 3544 4554 5564 6575
Age, years Age group, years
20 20
Androstenedione
Androstenedione nmol/l
Androstenedione nmol/l

15 15

10 10

5 5

0 0
20 30 40 50 60 70 80 1824 2534 3544 4554 5564 6575
Age, years Age group, years

Fig. 23.1 Relationship between age and individual androgens in women (Davison et al. 2005a) with permission. To convert nmol/l to ng/dl or
pmol/liter to pg/dl, divide by 0.0347; to convert mmol/liter to mg/dl divide by 0.027. Raw data are represented as scattergraphs with fitted
regression curves. In the box and whisker plots, the box represents the interquartile range (IQR), the line in the box is the median. The whiskers
extend to the upper and lower adjacent values. The upper adjacent value is defined as the largest data point  the 75th percentile 1.5
 IQR. The lower adjacent value is defined as the smallest data point  the 25th percentile  1.5  IQR. Outliers are any values beyond the
whiskers. DHEAS dehydroepiandrosterone sulfate.

497
Chapter 23: Testosterone use in women

testosterone. These effects may be amplified in older Taking into consideration that endogenous hor-
women whose overall androgen production is declin- mone levels may be poor indicators of their clinical
ing. Treatment with oral glucocorticosteroids results effects, various studies have attempted to delineate the
in ACTH suppression and, hence, reduced adrenal relationships between testosterone and clinical char-
androgen production (Abraham 1974). Total testos- acteristics including sexual function, well-being, car-
terone levels are significantly lower in premenopausal diovascular disease (CVD) risk, insulin resistance and
and postmenopausal women with hypopituitarism cognition.
due to loss of both ovarian and adrenal androgen
production (Miller et al. 2001; Zang and Davis 2008). 23.3.1 Testosterone and sexual function
In the premenopausal years, other pathophysiolo-
The relationships between androgens and sexual func-
gical states such as hypothalamic amenorrhea and
tion were explored in a cross-sectional study of 1423
hyperprolactinemia are characterized by low circulat-
non-healthcare seeking women, aged 18 to 75 years,
ing testosterone levels (Miller et al. 2007). Similarly,
randomly recruited from the community via the elect-
women with premature ovarian failure have lowered
oral roll, of whom 1021 completed a validated sexual
androgen levels (Kalantaridou et al. 2006).
function questionnaire (Davis et al. 2005). Total and
free testosterone levels were not related to sexual func-
23.3 Clinical associations with tion scores. However, women aged 45 years or more
with low sexual responsiveness had a greater likelihood
endogenous testosterone levels in of having a serum DHEAS value below the 10th centile
women: findings from observational for their age (odds ratio 3.9; 95% confidence interval
studies 1.549.81; p = 0.004). For women aged 18 to 44 years,
having low sexual desire, sexual arousal or sexual
Studies investigating relationships between endogen-
responsiveness was also associated with having a
ous testosterone levels and health outcomes are con-
DHEAS value below the 10th centile for their age
founded by a number of factors. Androgens undergo
(Davis et al. 2005). As the normal range for serum
substantial metabolism in extra-gonadal tissues (Labrie
DHEAS amongst young women is relatively large and
1991), such that circulating blood levels in women may
a significant proportion of women with low DHEAS
not accurately reflect tissue concentrations or tissue
do not have low sexual function, a cutoff level below
effects. Precise measurement of testosterone at very
which women can be said to be more likely to have low
low concentrations has been an ongoing challenge for
sexual function could not be identified.
clinical research in this field (Taieb et al. 2003). Fur-
There is also evidence that testosterone is asso-
thermore, as testosterone is an obligatory precursor for
ciated with vaginal health, with levels being inversely
estrogen production, it is difficult to differentiate the
associated with vaginal atrophy in postmenopausal
independent effects of testosterone versus estrogen for
women (Leiblum et al. 1983). Testosterone has vasodila-
pathophysiological outcomes. Also often disregarded is
tory effects (Worboys et al. 2001), enhancing vaginal
that studies evaluating testosterone action commonly
blood flow and lubrication with sexual arousal (Leiblum
report both total testosterone levels and the FAI. The
et al. 1983; Tuiten et al. 2000). These effects may be due to
FAI (total testosterone / SHBG  100) is believed, by
direct androgen actions or in part be due to estradiol
some, to reflect biologically active testosterone. Some-
biosynthesis from testosterone in the vascular bed (Har-
times the term bioavailable testosterone is loosely
ada et al. 1999). The clinical significance of this is that the
used when unbound testosterone has been estimated
complaint of vaginal dryness is usually attributed to estro-
by the FAI. SHBG levels vary substantially in women,
gen deficiency, whereas failure to lubricate with sexual
with endogenous levels inversely linked to central adi-
stimulation may require adequate androgen action.
posity and insulin levels (Nestler and Strauss 1991;
Goodman-Gruen and Barrett-Connor 1997; Randolph
et al. 2003). In the many studies in which total testos- 23.3.2 Testosterone and well-being
terone is not associated with the outcome of interest, Lowered well-being is a common complaint amongst
but SHBG and the FAI or bioavailable testosterone women at midlife. However, evidence that low testos-
are, the likelihood is that SHBG is driving the terone underpins diminished well-being is lacking.
association. In a study in which 1224 premenopausal and

498
Chapter 23: Testosterone use in women

postmenopausal women completed a validated been associated with lower levels of androgen precur-
psychological general well-being questionnaire, no sors and a higher estradiol-to-testosterone ratio
relationship between overall well-being and total or (Naessen et al. 2010). Furthermore, women with a
free testosterone was identified (Bell et al. 2006). total testosterone level in the lowest quintile were
observed to have the greatest risk for all-cause mor-
23.3.3 Testosterone and cardiovascular tality and incident CVD, independent of traditional
risk factors, over a 4.5-year follow-up period (Sievers
disease et al. 2010). Polycystic ovarian syndrome (PCOS) is a
Observational studies consistently demonstrate that common condition characterized by hyperinsulin-
both endogenous testosterone and SHBG are inversely emia and androgen excess. The largest observational
associated with increased CVD risk in women. In a study reporting the morbidity and mortality associ-
cross-sectional, community-based study exploring the ated with this condition did not find an increase in
relationships between androgens and CVD risk coronary heart disease morbidity or mortality in
markers, total testosterone was not associated with women with PCOS, although these women had more
circulating lipid levels or CRP when age, smoking, CVD risk factors (Wild et al. 2000).
alcohol, BMI and exercise were taken into account Taken together, observational studies indicate low,
(Bell et al. 2007). For premenopausal women, free not high testosterone is associated with CVD risk in
testosterone explained 1% of the variance in CRP. women.
SHBG contributed significantly to variations in
CRP, triglycerides and HDL-C in both pre- and post-
menopausal women. Similarly other researchers have 23.3.4 Testosterone and osteoporosis
found a strong inverse relationship between SHBG Testosterone appears to have skeletal effects, either via
and CRP when adjusting for other variables (Joffe the AR or as a precursor for estradiol production in
et al. 2006). bone. Testosterone levels have been positively correl-
The FAI, but not total testosterone, has been ated with BMD in premenopausal and postmenopau-
related to the metabolic syndrome and increased sal women (Slemenda et al. 1996; Rariy et al. 2011).
CVD risk (Golden et al. 2004; Sutton-Tyrrell et al. For women aged 67 years or more, total testosterone
2005). The FAI has been strongly associated with is positively associated with BMD at the lumbar spine
visceral fat in postmenopausal women, even after and hip, even after adjusting for estradiol, and free
adjusting for insulin resistance (Janssen et al. 2010). testosterone is positively associated with hip BMD,
The FAI and SHBG were interchangeable in their lean body mass, and body fat (Rariy et al. 2011).
strength of association with visceral fat, whereas total To investigate the relationship between testoster-
testosterone was not related to visceral fat. Thus one and hip fracture in postmenopausal women,
SHBG is the factor in the FAI associated with visceral 400 participants in the Womens Health Initiative
fat, not testosterone. Similarly, in young to middle- Observational Study who experienced their first
aged women, SHBG levels, but not total or free tes- non-pathological hip fracture (median follow-up,
tosterone, have been inversely associated with sub- seven years) were matched with 400 controls by age,
clinical CVD (Calderon-Margalit et al. 2010) and ethnicity and baseline blood draw date (Lee et al.
coronary artery calcium (Ouyang et al. 2009). Thus 2008). High endogenous testosterone was associated
SHBG, not testosterone, underpins the relationship with a significantly decreased risk of hip fracture,
between FAI and CVD risk factors in these studies. independent of SHBG, serum estradiol concentration
Endogenous testosterone has been positively asso- and other potential risk factors.
ciated with brachial artery flow-mediated dilatation, a In women with anorexia nervosa, total and free-
measure of endothelial function, in postmenopausal testosterone levels, but not estradiol, are positively
women (Montalcini et al. 2007). Free testosterone has associated with bone volume fraction and trabecular
been inversely associated with carotid intima-media thickness after controlling for BMI (Lawson et al.
thickness (Bernini et al. 1999), and total and free 2010). Androgen receptors have been demonstrated
testosterone are inversely associated with internal in human osteoblast-like cell lines, and androgens
carotid artery atherosclerosis (Debing et al. 2007). have been shown to directly stimulate bone cell pro-
Most recently, prevalent CVD in older women has liferation and differentiation (Kasperk et al. 1989).

499
Chapter 23: Testosterone use in women

23.3.5 Testosterone and cognitive exogenous testosterone improves the most commonly
reported sexual problems in women. These are sexual
function desire and arousal, pleasure and overall satisfaction
Testosterone has been reported to have neuroprotec- (Laumann et al. 1999; Hayes et al. 2008). For most
tive properties (Rosario et al. 2009). Higher endogen- women, these problems are part of a continuum of
ous testosterone levels in premenopausal women have the sexual experience, and are inextricably related.
been linked with better performance in tasks of spatial Recent studies evaluating the efficacy of testosterone
and mathematical ability (Gouchie and Kimura for the treatment of female sexual dysfunction (FSD)
1991); whilst in elderly women higher testosterone have required participants to fulfill the diagnostic
levels have been associated with superior performance criteria of hypoactive sexual desire disorder (HSDD),
on verbal fluency and verbal memory tasks (Wolf and which has been defined as persistent or recurrent
Kirschbaum 2002). Levels of testosterone in the deficiency or absence of sexual thoughts and fantasies
human female brain during the reproductive years and/or desire for, or receptivity for, sexual activity,
are several-fold greater than those of estradiol (Bixo causing personal distress or interpersonal difficulties
et al. 1995). Within the brain testosterone exhibits (American Psychiatric Association 1994).
neuroprotective effects including protection against The controversies regarding the use of testoster-
oxidative stress, serum deprivation-induced apoptosis one to treat HSDD are first whether this is a condition
and soluble beta amyloid (Ab) toxicity (Pike et al. that merits pharmacological intervention and second
2009). Although some of these effects of testosterone whether testosterone, as a pharmacological interven-
are blocked by aromatase inhibition and thus appear tion, is sufficiently effective and safe.
to be estrogen-mediated, protection against b tox-
icity appears to be AR-mediated (Pike et al. 2009).
There is also evidence that endogenous androgens
23.4.2 The need to treat hypoactive sexual
influence levels of Ab, possibly by an AR-dependent desire disorder
mechanism involving upregulation of the Ab-catabo- Population-based data show that most women believe
lizing enzyme neprilysin (Yao et al. 2008). In addition an active sex life is important for ones sense of well-
to having neuroprotective effects, testosterone has being, and that higher levels of physical pleasure in
positive effects on endothelial function (Montalcini sex are significantly associated with higher levels of
et al. 2007) and acts as a vasodilator (Worboys et al. emotional satisfaction. However, the prevalence of
2001), such that testosterone-induced vascular effects HSDD amongst postmenopausal women is of the
may contribute to neuroprotective effects. As mild order of 9 to 14%, with no differences between natural
cognitive impairment precedes the development of and surgically menopausal women (Shifren et al.
dementia by several years, and a parallel is seen 2008). The prevalence of this condition is consistent
between the fall of testosterone levels in women and across developed and undeveloped countries (Tung-
the rise in incidence of mild cognitive decline and phaisal et al. 1991; Dennerstein et al. 2006).
dementia, further exploration of this association is The impact of HSDD extends well beyond an
warranted. individuals sexual life. Hypoactive sexual desire dis-
order is associated with diminished health-related
23.4 Effects of testosterone: findings QoL in both naturally and surgically menopausal
women (Biddle et al. 2009). Women with HSDD are
from randomized, controlled trials more likely to be depressed and dissatisfied with their
23.4.1 The use of testosterone for the home life, and with the emotional and physical rela-
tionships they have with their sexual partner. The
treatment of female sexual dysfunction overall effect of HSDD on quality of life in women
There is no diagnostic lower limit for testosterone with HSDD is similar in magnitude to that seen in
or free testosterone that can be used to classify a adults with other common chronic conditions such as
woman as androgen deficient (Davis et al. 2005). diabetes and back pain (Biddle et al. 2009). Women in
Thus the use of testosterone therapy for women is the community who self-identify as being sexually
not based on an established link between symptoms dissatisfied have significantly lower well-being in
and biochemistry, but rather clinical evidence that comparison to women who self-identify as being

500
Chapter 23: Testosterone use in women

satisfied with their sexual life (Davison et al. 2009). A Cochrane Review concluded that the addition of
Aware of the importance of the sexual activity in the testosterone to postmenopausal estrogen therapy
relationship, women commonly continue to engage improves sexual function in postmenopausal women
in sexual activity despite experiencing little sexual (Somboonporn et al. 2005).
desire or pleasure, or dyspareunia (Manderson 2005; Research into the TTP followed the guidelines set
Davison et al. 2008; Avis et al. 2009). Overall, loss of out by the FDA. For evidence that testosterone ther-
sexual desire for many women leads to lowered well- apy was an effective treatment for HSDD, the FDA
being and increased anxiety and personal distress, required the primary outcome to be the frequency of
translating into relationship disharmony, often with satisfactory sexual events determined by the treated
an unavoidable negative ripple effect impacting other woman. Study participants were thus required to keep
family members. Just as individuals seek treatment a daily diary documenting each sexual event and
for other factors that impact negatively on their qual- stating whether it was satisfactory or not. Women
ity of life such as depression, women experiencing not in a monogamous ongoing relationship and
HSDD should be encouraged to discuss their prob- women found to be depressed or experiencing poor
lems with a healthcare provider. relationship satisfaction were excluded from these
recent large, randomized, placebo-controlled trials
(RCTs).
23.4.3 Efficacy of testosterone for As already stated, women who report being dis-
treatment of hypoactive sexual desire satisfied with their sexual life continue to engage in
disorder: findings from randomized, sexual activity, such that the total frequency of sexual
engagement is a poor measure of a womans sexual
controlled trials well-being (Manderson 2005). Further evidence for
The early studies demonstrating efficacy of testoster- this is provided by a recent study which found that
one in postmenopausal women with FSD were small postmenopausal women who self-identified as being
and involved the use of testosterone implants (Burger dissatisfied with their sexual well-being still recorded
et al. 1987) or injections (Sherwin and Gelfand 1987). a median of five sexual events per month (Davison
They were not selective for women fulfilling the cri- et al. 2008). Similarly, at baseline women recruited to
teria of HSDD. A single-blind study that compared a large, randomized, placebo-controlled trial of tes-
estradiol implants alone, versus estradiol plus testos- tosterone treatment for HSDD also reported on
terone implants, in postmenopausal women experi- average five sexual events per month, with only
encing lowered libido, found sustained benefits of the 50% of these events being considered by the women
addition of testosterone when women were assessed as sexually satisfying (Davis et al. 2008a). Therefore,
by a validated sexual function questionnaire every six evaluation of treatment efficacy needs to include
months over two years (Davis et al. 1995). Similar other aspects of sexual function important to the
benefits of oral methyltestosterone therapy were sub- woman rather than just a count of total activities.
sequently reported (Sarrel et al. 1998; Lobo et al. 2003; Hence various questionnaires have been developed
Warnock et al. 2005). However, concerns that oral and validated to assess sexual desire, arousal, plea-
methyltestosterone lowers HDL-C and may increase sure and other parameters of the female sexual
weight and visceral fat accumulation (Leao et al. 2006; response.
Somboonporn et al. 2005) have resulted in dimin- In two large studies of the TTP versus placebo in
ished interest in its use and the development of non- surgically menopausal women on oral estrogen therapy,
oral testosterone therapies for women. The most the increase in the number of satisfying sexual events
extensively investigated therapy has been the trans- per month was from three times a month to five times
dermal testosterone patch (TTP) which delivers 300 per month with active therapy (Buster et al. 2005; Simon
mg of testosterone per day. Although the TTP as a et al. 2005). There was an associated decline in personal
treatment for HSDD was studied first in surgically distress, a key component of HSDD, which decreased by
menopausal women using oral estrogen, it has 65% and 68% in each of these two large studies with
been shown to be efficacious in both naturally and TTP 300 mg/day, compared with 40% and 48% with
surgically menopausal women taking oral and non- placebo (Fig. 23.2). All domains of sexual function
oral estrogen or not using concurrent estrogen. (arousal, pleasure, orgasm, self-image, reduced concern,

501
Chapter 23: Testosterone use in women

(A) Fig. 23.2 Changes in four-week


Total satisfying sexual activity frequency of (A) total satisfying sexual
3
4-wk mean change from activity, (B) sexual desire score and (C)
2.5 personal distress score in postmenopausal
women treated with a testosterone patch
2 releasing 300 mg/day, 150 mg/day or
baseine

placebo over 24 weeks. Symbols: ,


1.5 testosterone; , placebo; *, p  0.05 vs.
1 placebo. (Simon et al. 2005, with
permission.)
0.5

0
0 4 8 12 16 20 24
Weeks

(B) Sexual desire

14
12
Mean change from
baseline (SEM)

10
8
6
4
2
0
0 4 8 12 24
Weeks

(C) Personal distress


0
Mean change from baseline

10

15

20

25

30
0 4 8 12 24
Weeks

and responsiveness) also improved to a significantly to be similar in postmenopausal women treated with
greater extent with testosterone than with placebo. Nat- transdermal estrogen and those not using any concur-
urally menopausal women with HSDD receiving a rent estrogen therapy in RCTs (Davis et al. 2006a;
stable dose of oral estrogen had a mean increase from 2008a; Panay et al. 2010).
baseline in their four-week frequency of satisfying The general emphasis has been on the develop-
sexual events with TTP 300 mg/day of 1.92 (73%) com- ment of testosterone as a treatment for postmeno-
pared with placebo (0.5, (19%)), accompanied by pausal women. But testosterone levels do not
increases in the other domains of sexual function decline with natural menopause. So, women who
assessed (Fig. 23.3), and decreased distress (Shifren have lowered testosterone in their early postmeno-
et al. 2006). Efficacy of TTP therapy has been shown pausal years would have had lowered testosterone in

502
Chapter 23: Testosterone use in women

Placebo
20 Testosterone

18
44%
16
Mean change from baseline (SEM)

55%
14

12
37%
25%
10 31%
48%
8 33%
29% 21%
6
12%
4
20% 13%
2 7%
7%
0
Decreased
Desire Arousal Orgasm Pleasure concerns Responsiveness Self-image
p 0.01 vs. placebo
p 0.001 vs. placebo
Fig. 23.3 Mean change from baseline in Profile of Female Sexual Function domain scores, by treatment group, at week 24 (intent-to-treat
population). In naturally menopausal women on oral estrogen with and without transdermal testosterone therapy. (Shifren et al. 2006, with
permission.)

their late reproductive years. Therefore, in patients Of note, when administered transdermally at doses
with HSDD, the argument for treating women in that restore levels of free testosterone to those seen in
their late reproductive years with testosterone is as healthy young women, the effects of testosterone on
strong as for treating naturally menopausal women sexual function do not become manifest until about six
with testosterone. Two randomized placebo-con- to eight weeks of treatment. Transdermal testosterone
trolled trials have also shown that transdermal tes- appears to be less effective for women with an elevated
tosterone improves the frequency of satisfactory SHBG level (> 160 pmol/l) (Shifren et al. 2006) or if
sexual events and other parameters of sexual well- used by women taking conjugated equine estrogens
being in older premenopausal women presenting (Procter and Gamble Pharmaceuticals 2004).
with loss of libido (Goldstat et al. 2003; Davis With respect to the local vaginal effects of testos-
et al. 2008b). Concern regarding the safety of tes- terone, there are some preliminary data that the
tosterone in women who have the potential to con- vaginal application of testosterone, 300 mg/d, may
ceive has limited its use in premenopausal women. alleviate dyspareunia in women with breast cancer,
However, restoration of testosterone levels to within without raising circulating blood levels (Witherby
the normal range for young healthy women would et al. 2011). This merits further evaluation.
not virilize a female fetus. Indeed, the rise in SHBG The mechanism of action of testosterone in terms
in early pregnancy protects the fetus from maternal of sexual function in women has not been extensively
androgen exposure. Treatment with transdermal researched. In postmenopausal women treated with a
testosterone also has been shown to significantly stable dose of transdermal estradiol, the effects of
improve sexual function in women with hypopitui- testosterone therapy were unaltered by the concurrent
tarism (Miller et al. 2006). administration of an aromatase inhibitor in an RCT,

503
Chapter 23: Testosterone use in women

indicating that the therapeutic effect of testosterone et al. 2005; Davis et al. 2008a). In women with docu-
on sexual desire and arousal is a consequence of direct mented congestive cardiac failure, treatment with the
androgen action, as opposed to the aromatization of TTP releasing 300 mg of testosterone per day was
testosterone to estradiol (Davis et al. 2006b). associated with significant functional improvements
Taken together, the available clinical evidence sup- assessed by peak oxygen consumption, distance
ports the efficacy of parenteral testosterone therapy as walked over the six-minute walking test, muscle
part of the management of lowered sexual desire and strength and insulin resistance, compared with pla-
arousal in women who have undergone a comprehen- cebo (Lellamo et al. 2010). None of the RCTs com-
sive clinical evaluation to exclude other causes. paring the transdermal testosterone therapy with
placebo have shown a difference in event rate for
any CVD outcome, including venous thromboem-
23.4.4 Effects of testosterone therapy on bolic events, versus placebo.
mood and well-being
Most large RCTs of testosterone versus placebo 23.4.6 Effects on bone and lean mass
have included assessments of well-being in their Small studies have shown that testosterone treatment
design. A significant improvement in well-being, will increase vertebral and hip BMD in postmenopau-
measured by a validated questionnaire, has only sal women (Davis et al. 1995; Watts et al. 1995). In
been observed in RCTs in which the study popula- women of reproductive years with hypopituitarism,
tion had a baseline well-being score below that of treatment with the TTP (300 mg daily) for 12 months
the general population (Shifren et al. 2000; Goldstat resulted in significant increases in BMD at the hip
et al. 2003; Davis et al. 2006a). Testosterone should and radius, and in lean body mass (Miller et al. 2006).
not be considered a treatment option for mood Similarly, oral methyltestosterone has been shown to
disorders in women. significantly increase BMD in women with Turner
syndrome, versus placebo (Zuckerman-Levin et al.
2009). Whether testosterone therapy will prevent frac-
23.4.5 Effects of testosterone therapy on tures has not been investigated.
cardiovascular function and cardiovascular Overall, studies evaluating the effects of testoster-
disease risk one on body composition in postmenopausal women
report increased lean mass and reduced body fat. In
Studies of testosterone administered by subcutaneous
postmenopausal women, testosterone implant ther-
implant (Davis et al. 1995), transdermal patch (Simon
apy over two years increased lean body mass as
et al. 2005; Davis et al. 2008a), spray (Davis et al.
assessed by DXA (Davis et al. 2000). Combined oral
2008b) or gel (Davis et al. 2006b) do not show
methyltestosterone with oral estrogen therapy results
any adverse effects in terms of altered lipid levels,
in increased lean mass, reduced percentage body
CRP, glycosylated hemoglobin (HbA1c) or insulin
fat and increased lower body strength compared to
sensitivity. However, HDL-C and apolipoprotein
estrogen therapy alone (Dobs et al. 2002). In women
A1 levels decrease significantly when oral methyltes-
with Turner syndrome, oral methyltestosterone
tosterone is administered with oral estrogen (Barrett-
increased total trunk lean body mass, whereas total
Connor et al. 1999; Chiuve et al. 2004). Combined
fat mass decreased and the visceral fat and visceral-to-
estrogen and methyltestosterone therapy is also asso-
subcutaneous fat ratio, as evaluated by abdominal CT,
ciated with reduced plasma concentrations of apoli-
did not change with testosterone compared with pla-
poprotein B, reduced LDL particle size, and increased
cebo (Zuckerman-Levin et al. 2009). In women with
total body LDL catabolism (Wagner et al. 1996).
hypopituitarism, treatment with the TTP did not
Similarly, oral testosterone undecanoate adversely
affect intra-abdominal or subcutaneous fat mass as
affects lipoproteins and increases insulin resistance
measured by CT (Miller et al. 2006).
(Zang et al. 2006).
SHBG is a strong independent marker of insulin
resistance and the risk of type 2 diabetes. Levels of 23.4.7 Effects of testosterone on cognition
SHBG do not change with transdermal testosterone High-quality RCT data for the effects of testosterone
therapy in studies over 26 to 52 weeks (Braunstein on cognitive performance in women are lacking.

504
Chapter 23: Testosterone use in women

Some studies have reported effects of a single dose 23.5 Safety of testosterone therapy
(Aleman et al. 2004) or have been of short duration
(Kocoska-Maras et al. 2011), such that the clinical for women
significance of the findings is unclear. Other longer 23.5.1 Testosterone and adverse
and larger studies mostly indicate a favorable effect of
testosterone on cognitive performance, but the find- androgenic effects
ings are not consistent and all have methodological The potential masculinizing effects of androgen ther-
limitations. A significant improvement in immediate apy include development of acne, hirsutism,
and delayed verbal memory using the California deepening of the voice and androgenic alopecia.
Verbal Learning Test was observed in an open-label These effects are dose related and are uncommon if
study of transdermal testosterone in postmenopausal supraphysiological hormone levels are avoided. Com-
women on estrogen therapy (Shah et al. 2006); pared with placebo therapy, women treated with
whereas a negative effect on immediate but not transdermal testosterone in various studies report a
delayed verbal memory was observed over 24 weeks higher rate of androgenic adverse events, mainly
with testosterone undecanoate as an oral therapy attributable to increased hair growth (Fig. 23.4).
(Moller et al. 2010). In a study of oral estrogen alone However, withdrawal from research studies due to
or in combination with methyltestosterone, women androgenic adverse events has not been seen to be
receiving estrogen and methyltestosterone maintained greater in women treated with testosterone (Davis
a steady level of performance on the Building et al. 2008a). Hirsutism, androgenic alopecia and/or
Memory task, whereas those receiving estrogen alone acne are relatively strong contradictions to androgen
showed a decrease in performance (Wisniewski et al. therapy.
2002). An important consideration in terms of meth-
odology is that all of the published studies of the
effects of testosterone on cognitive performance have 23.5.2 Testosterone and breast cancer risk
employed tests developed for cognitively impaired The main concern of regulators and the community is
individuals, which are not sensitive to small changes whether testosterone therapy may influence breast
in cognitive performance within the normal range for cancer risk. Testosterone treatment results in growth-
healthy women. Findings are also confounded by the inhibitory and apoptotic effects in some, but not all,
well-documented practice effects observed when nor- breast cancer cell lines, and, in rodent breast cancer
mally functioning individuals are exposed to the same models, despite the potential for testosterone to be
test materials within a brief time interval. Studies aromatized to estradiol, testosterone action is generally
using more sophisticated measures of cognitive per- antiproliferative and proapoptotic (Somboonporn and
formance developed for cognitively intact individuals, Davis 2004). It is of note that from the 1940s through
that are resistant to practice effects, are needed. until the 1970s, testosterone was used in the treatment
of breast cancer (Adair and Herrmann 1946; Rieche
and Wolff 1975).
23.4.8 Use of testosterone for treatment of For premenopausal women, data pertaining to
vulval lichen sclerosis testosterone and breast cancer risk is limited to find-
Vulval lichen sclerosis is a chronic skin condition ings from observational studies of endogenous hor-
characterized by pruritus, irritation, burning, dyspar- mone levels, as testosterone therapy is rare in
eunia and tearing. It most commonly occurs after the premenopausal women, and the only RCTs under-
fifth decade. The typical appearance is porcelain- taken have been too small to provide useful data.
white papules and plaques, often with areas of The significance of the available data is limited by
ecchymosis or purpura affecting the vulval tissue. failure to take into account timing of blood drawing
The mainstay of treatment for this condition is in relation to the menstrual cycle or time of day,
high-dose topical steroid cream, usually clobetasol imprecision of assays used, and failure to take into
propionate. Although testosterone cream has been account estradiol levels. Most studies do not demon-
recommended in the past, the findings have been strate an association between testosterone and breast
inconsistent, and adverse androgenic effects have cancer risk in premenopausal women (Somboonporn
been reported (ACOG 2008). and Davis 2004). One prospective, case control study

505
Chapter 23: Testosterone use in women

Percentage of Fig. 23.4 Effects of different doses of


each treatment the transdermal testosterone patch on
group affected androgenic events over 12 months in
postmenopausal women not using
25 concurrent estrogen therapy. Adapted
from Davis et al. (2008a). See plate section
for color version.
20

15
Placebo
10 150 g TTP
300 g TTP
5

0
Acne Increased hair Alopecia Voice change
growth

has reported a relationship between testosterone no use of hormone therapy, was reported (adjusted
and increased risk of subsequent breast cancer, hazard ratio:1.42; 95% CI: 0.952.11) in the setting of
although estradiol levels were not taken into account 49% of users also being progestin users, compared
in the analysis (Dorgan et al. 2010). Despite PCOS with 11% of non-hormone users being past estro-
being characterized by chronic estradiol and estrone gen-progestin users (Ness et al. 2009). A third, large,
exposure and androgen excess, women with PCOS do case control study of women aged 5064 years
not have an increased risk of breast cancer (Coulam reported no effect of methyltestosterone use on breast
et al. 1983; Gammon and Thompson 1991; Anderson cancer risk (Jick et al. 2009).
et al. 1997). Cohort studies of women treated with either tes-
The older observational studies of endogenous tosterone implants or transdermal testosterone do not
testosterone and breast cancer risk in postmenopausal show an increase in risk with testosterone therapy
women are also limited by assay imprecision (Som- (Dimitrakakis et al. 2004; Davis et al. 2009a). There
boonporn and Davis 2004). More recent studies has been no evidence of an increased risk in associ-
undertaken within the NSABP Cancer Prevention ation with duration of exposure and no evidence of
Trial (Beattie et al. 2006) and the Nurses Health altered risk in past users of testosterone therapy in the
Study (Danforth et al. 2010) showed no significant above-mentioned studies. Testosterone therapy does
association between breast cancer risk and any of the not alter mammographic density over 12 months
androgens measured. (Davis et al. 2009b). No RCT has been of sufficient
Observational studies of women using methyltes- size or duration to provide definitive evaluable data
tosterone have produced mixed results. The Nurses for the impact of testosterone on breast cancer risk.
Health Study reported an increase in risk for current
but not past users of methyltestosterone therapy up to 23.5.3 Testosterone and endometrial
2002 (Tamimi et al. 2006). This was an older cohort
(mean age 61.5 years). There were 29 cases of breast cancer risk
cancer in estrogen plus testosterone users and 3 cases Endometrial cancer is more common amongst
in testosterone-only users in 5628 women-years of women with obesity (Lindemann et al. 2008), diabetes
follow-up. Testosterone users were younger, leaner, (Lindemann et al. 2008) and polycystic ovarian syn-
more likely to have benign breast disease (55%) and drome (PCOS) (Wild et al. 2000) and/or prolonged
consumed more alcohol than controls, and the indi- unopposed exposure to estrogen (Antunes et al. 1979)
cations for testosterone therapy were not known. In and agents which are estrogen-receptor agonists in
another study no significant effect on breast cancer the endometrium, such as tamoxifen (Thurlimann
risk of estrogen plus methyltestosterone use, versus et al. 2005). There has been no association between

506
Chapter 23: Testosterone use in women

free testosterone and endometrial cancer risk after This form of sexual activity was most common
adjustment for estrone and estradiol levels (Allen amongst single, widowed and divorced women.
et al. 2008). Data pertaining to the effects of treatment The importance of this observation is that the
with testosterone on the endometrium are limited. desire to experience sexual well-being is not limited
Tissue samples from female-to-male transsexuals to women in relationships, and that sexual desire
treated with high-dose testosterone for, on average, and capacity to be aroused is an important consid-
three years prior to hysterectomy show inactive endo- eration for all women. Although most of the pub-
metria, with expression of Ki-67 in the glands, lished studies of testosterone have required
stroma, and glands and stroma together being signifi- participants to be in established monogamous rela-
cantly lower than for controls (Perrone et al. 2009). tionships, this should not be a requirement for
Transdermal testosterone is associated with endomet- consideration of testosterone therapy in clinical
rial atrophy (Zang et al. 2007; Davis et al. 2008a), and practice.
no cases of endometrial hyperplasia or carcinoma An array of physical, psychological, cultural and
have been diagnosed in clinical trials of testosterone relationship factors influence sexual well-being and
therapy. Taken together, the available data do not sexual function. Hence women presenting with the
indicate that treatment with testosterone significantly complaint of diminished sexual interest with or with-
influences endometrial cancer risk. out impaired sexual responsiveness need a general
psychological, physical and social health assessment.
Commonly contributing factors include stress,
23.5.4 Other safety concerns fatigue, relationship issues, depression and medica-
None of the RCTs of testosterone implants or trans- tion side-effects. Although identification of such
dermal testosterone patch or gel have reported liver factors does not exclude a woman from consideration
toxicity, polycythemia or other adverse metabolic for treatment with testosterone, such factors need to
effects. Sleep apnea has also not been reported in be managed. For example, relationship issues are not
women receiving physiological doses of testosterone. uncommonly a consequence of a womans dimin-
ished desire to engage in sexual activity, rather than
23.6 Appropriate candidates for the primary cause.
Loss of sexual desire is not uncommon amongst
testosterone therapy women using the combined oral contraceptive pill.
23.6.1 Identifying women likely to benefit Simply switching to another form of contraception
will often improve sexual well-being in younger
from testosterone therapy women. Concurrent use of the combined oral contra-
Overall, women who present with HSDD from the ceptive pill did not appear to impede response to
late reproductive years and beyond are potential can- testosterone therapy (Davis et al. 2008b).
didates for testosterone therapy. Physicians should The use of antidepressants is commonly associ-
specifically consider women who have experienced ated with impaired sexual function and may reflect
premature ovarian failure or surgical menopause, or either inadequate treatment of depression or a drug
who have adrenal insufficiency or hypopituitarism, as side-effect (Werneke et al. 2006). Unfortunately
having a high likelihood of HSDD, as these are con- research into the adverse effects of antidepressants
ditions characterized by low androgen production. and many other drugs on sexual function in women
Consideration should also be given to women who is sorely lacking. Women who present primarily with
have experienced iatrogenic ovarian failure secondary arousal disorder and inability to achieve orgasm, but
to chemotherapy, radiotherapy or chemical ovarian no significant loss of libido, in association with anti-
suppression. depressant therapy, may respond well to phospho-
A recent study by Avis and others (Avis et al. diesterase type 5 inhibitor therapy (Nurnberg et al.
2009) provides a broad insight into sexual activity 2008). Whether testosterone therapy will benefit
across the menopause. In this large cohort, 50% of women with antidepressant-associated FSD is yet to
postmenopausal women reported engaging in mas- be determined, as most studies of testosterone in
turbation, with 17% of women reporting doing so women have excluded women with depression and
once to twice a month and 10% at least once a week. users of antidepressants.

507
Chapter 23: Testosterone use in women

Table 23.1 Assessment of women presenting with sexual Table 23.2 Contraindications to testosterone therapy in
dysfunction women

1. Medical history Pregnancy and lactation


 Current circumstances Current use of antiandrogen therapy
 Relationship issues: consider referral for relationship Troublesome acne or hirsutism
counseling
Hormone-dependent malignancy current or past
 Employment and social engagement
SHBG level below the lower limit of normal
 Sexual experiences and sexual health knowledge,
Free testosterone level in the mid-normal range for
including individual and partners understanding of
young women and above
anatomy and sexuality and whether the women is
experiencing sexual stimulation: consider referral
for sexual counseling
 Health history, medications, drug use
Laboratory investigations should only be performed
2. Medical examination, including genital and pelvic as indicated and might include exclusion of factors
examination, particularly for loss of sensitivity or
causing fatigue (iron deficiency, thyroid dysfunction
pain disorders
and hyperglycemia). There is no point in measuring
3. Identify and manage health-related factors reproductive hormones in women taking the com-
a. Mental health bined oral contraceptive pill, as it suppresses estradiol
and testosterone production and increases SHBG.
i. Body image and self-esteem
As there is no level of testosterone, total or free,
ii. Experience of sexual abuse/trauma below which a woman can be diagnosed as being
iii. Negative attitudes, inhibitions and anxieties, androgen deficient, the measurement of testosterone
cultural and religious beliefs should not be used to indicate which women merit
b. Physical health
treatment. However, a testosterone level should be
measured prior to therapy primarily to exclude
c. Medication side-effects, particularly antidepressants women who may be at risk of side-effects if treated.
and antipsychotics Total testosterone is notoriously difficult to measure
d. Partners mental and physical health at lower levels, as seen in women, with sensitivity and
4. Consider testosterone therapy precision (Davison et al. 2005b). Clinicians interested
in this field of therapeutics should discuss with their
pathology service the sensitivity and precision of the
23.6.2 Assessment of women presenting assays being employed. Ideally total testosterone (pro-
tein-bound and free testosterone) should be measured
with low sexual desire as well as SHBG, and from this free testosterone can
It is important that each woman be assessed in the be reliably estimated by the pathology service using
context of her personal circumstances, partnership the Sodergard equation (Sodergard et al. 1982). The
status, sexual experiences and cultural expectations measurement of free testosterone in women by direct
(Table 23.1). It should not be assumed that a woman kit assays is generally completely unreliable (Herold
is experiencing adequate sexual stimulation, both and Fitzgerald 2003). Abnormal levels can be found
physical and emotional, such that this does not need when androgen excess is not expected.
to be explored. Referral to either a psychologist or Contraindications to testosterone therapy are pri-
sexual counselor is often appropriate. Medical history marily related to increased risk of worsening existing
needs to include general medical, psychosocial and acne, hirsutism or alopecia, pregnancy and lactation,
urogynecological details and the use of all medica- and unknown effects on hormone-dependent cancer
tions, both prescription and non-prescription. (Table 23.2). A low SHBG will enable more rapid
A medical examination should include a full genital clearance of administered testosterone, and hence
and pelvic assessment, including assessment of sensi- increased risk of androgenic side-effects. Similarly
tivity in women with loss of sensitivity or sexual pain. women with a free testosterone level in the

508
Chapter 23: Testosterone use in women

high-normal or above normal range for healthy elevated testosterone levels and androgenic effects,
young premenopausal women (Davison et al. 2005a) this practice cannot be condoned. Based on pre-
are at greater risk of overtreatment. scription and physician survey data, the number of
physicians prescribing compounded testosterone
formulations is increasing (Snabes and Simes
23.7 Treatment options 2009). Caution is urged, however, as there are no
Available data indicate that the most physiological published pharmacokinetic or safety data or effi-
mode of delivery of testosterone is parenteral, and cacy studies to validate this method of
primarily as a transdermal formulation. To date the administration.
testosterone patch has only been approved for the
treatment of surgically menopausal women using
concurrent estrogen in European Union countries. 23.8 Conclusions
A transdermal testosterone cream is available in Testosterone has been widely used by women as
Australia, with small studies showing efficacy an unapproved therapy for decades. In appropri-
in premenopausal and postmenopausal women ate doses, it is effective for the treatment of
(Goldstat et al. 2003; El-Hage et al. 2007). Increased low sexual desire in women approaching and
hair growth over the application site is a problem for following menopause. Large well-controlled trials
some women. have shown testosterone improves sexual desire,
In some countries testosterone is available as tes- arousal, orgasm frequency, pleasure and overall
tosterone pellets implanted subcutaneously under satisfaction and decreases distress associated with
local anesthetics. Most commonly a dose of 50 mg is FSD. It should also be considered in the manage-
used (Davis et al. 1995). These implants usually ment of women with premature ovarian failure
remain effective for periods of four to six months, and hypopituitarism. Possible beneficial effects of
although there is considerable intraindividual and testosterone on fracture risk, cognitive function
interindividual variation in their absorption and deg- and cardiovascular function require further
radation. Therefore, repeat implantation should not investigation.
be undertaken without confirmation that total testos- Excessive therapy will clearly result in undesirable
terone corrected for SHBG, or free testosterone, has androgenic effects such as hirsutism and acne,
fallen back into the lower quartile of the normal although this is not common when treatment is
female range. Otherwise, the risk of adverse andro- aimed at achieving testosterone levels in the female
genic effects is increased with the insertion of a new range. The capacity for both the breast and endomet-
implant. rium to aromatize testosterone to estradiol underlies
Other parenteral modes of delivery of testosterone concern regarding the potential for testosterone ther-
under development include a transdermal metered- apy to increase the risk of breast and endometrial
dose testosterone spray, a transdermal gel and an cancer. No RCT has been sufficiently large or of
intranasal testosterone gel. sufficient duration to clarify the effects of exogenous
Neither oral testosterone undecanoate nor methyl- testosterone on cancer risk, although cumulative data
testosterone can be recommended for women, as from the RCTs conducted to date do not indicate an
both may adversely affects lipids, and testosterone increase in risk.
undecanoate may induce insulin resistance. Some Availability of approved testosterone therapies for
clinicians undertake a clinical trial of im injections of women is limited to a few countries, and, in many of
testosterone esters 50100 mg. This may or may not these, approved use is limited to specific populations.
result in a clinical response over one to two weeks. Thus many clinicians are forced to resort to adapting
A positive response supports the initiation of products approved for men or the prescription of
longer-term therapy. However, as peak levels are compounded products. However, the extensive off-
very supraphysiological, testosterone esters should label prescribing of testosterone products for women
not be considered a long-term treatment option. raises serious safety concerns, and together with the
There is extensive off-label prescribing of tes- evidence for the negative impact of FSD on quality of
tosterone products, formulated for men, to treat life, highlights the need for an approved testosterone
women. As this approach may result in grossly formulation for women.

509
Chapter 23: Testosterone use in women

23.9 Key messages


milieu, being suppressed in the setting of insulin
 Levels of testosterone and DHT decline during resistance, hepatic steatosis and obesity.
the reproductive years, but do not change  In circumstances in which total testosterone is
acutely across natural menopause. not associated with health outcomes, such as
 There is no cutoff level for testosterone or free cardiovascular disease, but free testosterone is,
testosterone below which women can be labeled this is usually a consequence of low SHBG, not a
as androgen deficient. direct androgen effect.
 Transdermal testosterone therapy improves  Whether testosterone treatment increases the
sexual desire, arousal and global sexual risk of breast cancer is still debated. There is no
satisfaction in both premenopausal and evidence that past users of testosterone have an
postmenopausal women presenting with low increased risk of breast cancer and there are no
sexual desire which causes them concern. signals that transdermal testosterone modifies
 Free testosterone levels are dependent on SHBG, breast cancer risk. Studies further evaluating this
which is highly responsive to the metabolic are underway.

23.10 References hormones and endometrial cancer


risk in women in the European
estrogen-androgen and conjugated
estrogens in surgically menopausal
Abraham GE (1974) Ovarian and adrenal Prospective Investigation into women. Effects on bone mineral
contribution to peripheral androgens Cancer and Nutrition (EPIC). density, symptoms and lipid profiles.
during the menstrual cycle. J Clin Endocr Relat Cancer 15:485497 J Reprod Med 44:10121020
Endocrinol Metab 39:340346
American Psychiatric Association Beattie MS, Costantino JP, Cummings
ACOG (American College of (1994) Diagnostic and Statistical SR, Wickerham DL, Vogel VG,
Obstetrics and Gynecology) (2008) Manual of Mental Disorders. Dowsett M, Folkerd EJ, Willett WC,
ACOG Practice Bulletin No. 93: American Psychiatric Press, Wolmark N, Hankinson SE (2006)
diagnosis and management of Washington, DC Endogenous sex hormones, breast
vulvar skin disorders. Obstet cancer risk, and tamoxifen response:
Anderson KE, Sellers TA, Chen PL,
Gynecol 111:12431253 an ancillary study in the NSABP
Rich SS, Hong CP, Folsom AR
Adair FE, Herrmann JB (1946) The use (1997) Association of Stein- Breast Cancer Prevention Trial
of testosterone propionate in the Leventhal syndrome with the (P-1). J Natl Cancer Inst 98:110115
treatment of advanced carcinoma of incidence of postmenopausal breast Bell R, Donath S, Davison S, Davis S
the breast. Ann Surg 123:10231035 carcinoma in a large prospective (2006) Endogenous androgen levels
study of women in Iowa. Cancer and wellbeing: differences between
Aleman A, Bronk E, Kessels RP,
Koppeschaar HP, van Honk J (2004) 79:494499 pre- and postmenopausal women.
Antunes C, Strolley P, Rosenshiein N, Menopause 13:6571
A single administration of testosterone
improves visuospatial ability in young Davies J, Tonascia J, Brown C, Bell RJ, Davison SL, Papalia MA,
women. Psychoneuroendocrinology Burnett L, Rutledge A, McKenzie DP, Davis SR (2007)
29:612617 Pokempner M, Garcia R (1979) Endogenous androgen levels and
Endometrial cancer and estrogen cardiovascular risk profile in
Allen NE, Key TJ, Dossus L, Rinaldi S, women across the adult life span.
use. Report of a large case-control
Cust A, Lukanova A, Peeters PH, Menopause 14:630638
study. N Eng J Med 300:913
Onland-Moret NC, Lahmann PH,
Berrino F, Panico S, Larranaga N, Avis NE, Brockwell S, Randolph JF Jr, Bernini G, Sgro M, Moretti A,
Pera G, Tormo MJ, Sanchez MJ, Shen S, Cain VS, Ory M, Argenio GF, Barlascini CO, Cristofani
Ramon Quiros J, Ardanaz E, Greendale GA (2009) Longitudinal R, Salvetti A (1999) Endogenous
Tjonneland A, Olsen A, Chang- changes in sexual functioning as androgens and carotid intimal-medial
Claude J, Linseisen J, Schulz M, women transition through thickness in women. J Clin Endocrinol
Boeing H, Lundin E, Palli D, menopause: results from the Study Metab 84:20082012
Overvad K, Clavel-Chapelon F, of Womens Health Across the Biddle AK, West SL, DAloisio AA,
Boutron-Ruault MC, Bingham S, Nation. Menopause 16:442452 Wheeler SB, Borisov NN, Thorp J
Khaw KT, Bueno-de-Mesquita HB, Barrett-Connor E, Young R, (2009) Hypoactive sexual desire
Trichopoulou A, Trichopoulos D, Notelovitz M, Sullivan J, Wiita B, disorder in postmenopausal
Naska A, Tumino R, Riboli E, Yang HM, Nolan J (1999) A two- women: quality of life and health
Kaaks R (2008) Endogenous sex year, double-blind comparison of burden. Value Health 12:763772

510
Chapter 23: Testosterone use in women

Bixo M, Backstrom T, Winblad B, Chiuve SE, Martin LA, Campos H, randomized, placebo-controlled
Andersson A (1995) Estradiol and Sacks FM (2004) Effect of the trial. Menopause 13:387396
testosterone in specific regions of combination of methyltestosterone Davis SR, Goldstat R, Papalia MA,
the human female brain in different and esterified estrogens compared Shah SM, Kulkarni J, Donath S,
endocrine states. J Steroid Biochem with esterified estrogens alone on Bell R (2006b) Effects of aromatase
Mol Biol 55:297303 apolipoprotein CIII and other inhibition on sexual function and
apolipoproteins in very low density, wellbeing in postmenopausal
Braunstein GD, Sundwall DA, Katz M,
low density, and high density women treated with testosterone: a
Shifren JL, Buster JE, Simon JA,
lipoproteins in surgically randomized placebo-controlled
Bachman G, Aguirre OA, Lucas JD,
postmenopausal women. J Clin trial. Menopause 13:3745
Rodenberg C, Buch A, Watts NB
Endocrinol Metab 89:22072213
(2005) Safety and efficacy of a Davis SR, Moreau M, Kroll R,
testosterone patch for the treatment Coulam CB, Annegers JF, Kranz JS Bouchard C, Panay N, Gass M,
of hypoactive sexual desire disorder (1983) Chronic anovulation Braunstein GD, Linden-Hirschberg
in surgically menopausal women: syndrome and associated neoplasia. A, Rodenberg C, Pack S, Koch H,
a randomized, placebo-controlled Obstet Gynecol 61:403407 Moufarage A, Studd J (2008a)
trial. Arch Intern Med Couzinet B, Meduri G, Lecce M, Testosterone for low libido in
165:15821589 Young J, Brailly S, Loosfelt H, menopausal women not taking
Burger HG, Hailes J, Nelson J, Milgrom E, Schaison G (2001) The estrogen therapy. N Eng J Med
Menelaus M (1987) Effect of post menopausal ovary is not a 359:20052017
combined implants of estradiol and major androgen-producing gland. Davis SR, Papalia MA, Norman RJ,
testosterone on libido in J Clin Endocrinol Metab ONeill S, Redelman M,
postmenopausal women. BMJ 86:50605065 Williamson M, Stuckey BGA,
294:936937 Danforth KN, Eliassen AH, Wlodarczyk J, Gardner K,
Burger HG, Dudley EC, Hopper DL Tworoger SS, Missmer SA, Barbieri Humberstone A (2008b) Safety and
(1995) The endocrinology of the RL, Rosner BA, Colditz GA, efficacy of a testosterone metered-
menopausal transition: a cross- Hankinson SE (2010) The dose transdermal spray for
sectional study of a population- association of plasma androgen treatment of decreased sexual
based sample. J Clin Endocrinol levels with breast, ovarian and satisfaction in premenopausal
Metab 80:35373545 endometrial cancer risk factors women: a placebo-controlled
among postmenopausal women. Int randomized, dose-ranging study.
Buster JE, Kingsberg SA, Aguirre O, J Cancer 126:199207 Annals Internal Med 148:569577
Brown C, Breaux JG, Buch A,
Rodenberg C, Wekseman K, Davis SR, McCloud PI, Strauss BJG, Davis SR, Wolfe R, Farrugia H,
Casson P (2005) Testosterone patch Burger HG (1995) Testosterone Ferdinand A, Bell RJ (2009a) The
for low sexual desire in surgically enhances estradiols effects on Incidence of invasive breast cancer
menopausal women: a postmenopausal bone density and among women prescribed
randomized trial. Obstet Gynecol sexuality. Maturitas 21:227236 testosterone for low libido. J Sex
105:944952 Davis SR, Walker KZ, Strauss BJ (2000) Med 6:18501856
Effects of estradiol with and without Davis SR, Hirschberg AL, Wagner LK,
Calderon-Margalit R, Schwartz SM,
testosterone on body composition Lodhi I, von Schoultz B (2009b) The
Wellons MF, Lewis CE, Daviglus
and relationships with lipids in effect of transdermal testosterone
ML, Schreiner PJ, Williams OD,
post-menopausal women. on mammographic density in
Sternfeld B, Carr JJ, OLeary DH,
Menopause 7:395401 postmenopausal women not
Sidney S, Friedlander Y,
Davis SR, Davison SL, Donath S, Bell RJ receiving systemic estrogen therapy.
Siscovick DS (2010) Prospective
(2005) Circulating androgen levels J Clin Endocrinol Metab
association of serum androgens and
and self-reported sexual function in 94:49074913
sex hormone-binding globulin with
subclinical cardiovascular disease in women. JAMA 294:9196 Davison SL, Bell R, Donath S,
young adult women: the Coronary Davis SR, van der Mooren MJ, van Montalto JG, Davis SR (2005a)
Artery Risk Development in Young Lunsen RHW, Lopes P, Rees M, Androgen levels in adult females:
Adults womens study. J Clin Moufarege A, Rodenberg C, changes with age, menopause, and
Endocrinol Metab 95:44244431 Buch A, Purdie D (2006a) The oophorectomy. J Clin Endocrinol
efficacy and safety of a testosterone Metab 90:38473853
Campbell S, Whitehead M (1977)
Oestrogen therapy and the patch for the treatment of Davison SL, Bell R, Montalto JG,
menopausal syndrome. Clin Obstet hypoactive sexual desire disorder in Sikaris K, Donath S, Stanczyk FZ,
Gynaecol 4:3147 surgically menopausal women: a Davis SR (2005b) Measurement of

511
Chapter 23: Testosterone use in women

total testosterone in women: endogenous steroids to both Haning JRV, Hackett R, Flood CA,
comparison of a direct testosterone-binding globulin and Loughlin JS, Zhao QY, Longcope C
radioimmunoassay versus cortico-steroid-binding globulin in (1993) Plasma
radioimmunoassay after organic human plasma. J Clin Endocrinol dehydroepiandrosterone sulfate
solvent extraction and celite column Metab 53:5868 serves as a prehormone for 48% of
partition chromatography. Fertil Edmunds SE, Stubbs AP, Santos AA, follicular fluid testosterone during
Steril 84:16981704 Wilkinson ML (1990) Estrogen and treatment with menotropins. J Clin
Davison SL, Bell RJ, LaChina M, androgen regulation of sex hormone Endocrinol Metab 76:13011307
Holden SL, Davis SR (2008) Sexual binding globulin secretion by a Harada N, Sasano H, Murakami H,
function in well women: human liver cell line. J Steroid Ohkuma T, Nagura H, Takagi Y
stratification by sexual satisfaction, Biochem Mol Biol 37:733739 (1999) Localized expression of
hormone use, and menopause El-Hage G, Eden JA, Manga RZ (2007) aromatase in human vascular
status. J Sex Med 5:12141222 A double-blind, randomized, tissues. Circ Res 84:12851291
Davison SL, Bell RJ, La China M, placebo-controlled trial of the effect Hayes RD, Dennerstein L, Bennett CM,
Holden SL, Davis SR (2009) The of testosterone cream on the sexual Fairley CK (2008) What is the
relationship between self-reported motivation of menopausal true prevalence of female sexual
sexual satisfaction and general hysterectomized women with dysfunctions and does the way we
wellbeing in women. J Sex Med hypoactive sexual desire disorder. assess these conditions have an
6:26902697 Climacteric 10:335343 impact? J Sex Med 5:777787
Debing E, Peeters E, Duquet W, Fogle RH, Stanczyk FZ, Zhang X, Herold DA, Fitzgerald RL (2003)
Poppe K, Velkeniers B, Van den Paulson RJ (2007) Ovarian Immunoassays for testosterone in
Brande P (2007) Endogenous sex androgen production in women: better than a guess? Clin
hormone levels in postmenopausal postmenopausal women. J Clin Chem 49:12501251
women undergoing carotid artery Endocrinol Metab 92:30403043 Horton R, Hawks D, Lobo R (1982)
endarterectomy. Eur J Endocrinol Gammon MD, Thompson WD (1991) 3 alpha, 17 beta-androstanediol
156:687693 Polycystic ovaries and the risk of glucuronide in plasma. A marker of
Dennerstein L, Koochaki P, Barton I, breast cancer. Am J Epidemiol androgen action in idiopathic
Graziottin A (2006) Hypoactive 134:818824 hirsutism. J Clin Invest 69:
sexual desire disorder in Golden SH, Ding J, Szklo M, 12031206
menopausal women: a survey of Schmidt MI, Duncan BB, Dobs A Janssen I, Powell LH, Kazlauskaite R,
western European women. J Sex (2004) Glucose and insulin Dugan SA (2010) Testosterone and
Med 3:212222 components of the metabolic visceral fat in midlife women: the
Dimitrakakis C, Jones R, Liu A, syndrome are associated with Study of Womens Health Across
Bondy CA (2004) Breast cancer hyperandrogenism in the Nation (SWAN) fat patterning
incidence in postmenopausal postmenopausal women: the study. Obesity (Silver Spring)
women using testosterone in atherosclerosis risk in communities 18:604610
addition to usual hormone therapy. study. Am J Epidemiol 160:540548 Jick SS, Hagberg KW, Kaye JA, Jick H
Menopause 11:531535 Goldstat R, Briganti E, Tran J, Wolfe R, (2009) Postmenopausal estrogen-
Dobs AS, Nguyen T, Pace C, Roberts C Davis S (2003) Transdermal containing hormone therapy and
(2002) Differential effects of oral testosterone improves mood, well the risk of breast cancer. Obstet
estrogen versus estrogen-androgen being and sexual function in Gynecol 113:7480
replacement therapy on body premenopausal women. Menopause Joffe HV, Ridker PM, Manson JE,
composition in postmenopausal 10:390398 Cook NR, Buring JE, Rexrode KM
women. J Clin Endocrinol Metab Goodman-Gruen D, Barrett-Connor E (2006) Sex hormone-binding
87:15091516 (1997) Sex hormone-binding globulin and serum testosterone are
Dorgan JF, Stanczyk FZ, Kahle LL, globulin and glucose tolerance in inversely associated with C-reactive
Brinton LA (2010) Prospective case- postmenopausal women. The protein levels in postmenopausal
control study of premenopausal Rancho Bernardo Study. Diabetes women at high risk for
serum estradiol and testosterone Care 20:645649 cardiovascular disease. Ann
levels and breast cancer risk. Breast Gouchie C, Kimura D (1991) The Epidemiol 16:105112
Cancer Res 12:R98 relationship between testosterone Judd HL, Judd GE, Lucas WE, Yen SSC
Dunn JF, Nisula BC, Rodboard D levels and cognitive ability patterns. (1974) Endocrine function of the
(1981) Transport of steroid Psychoneuroendocrinology postmenopausal ovary.
hormones. Binding of 21 16:323334 Concentrations of androgens and

512
Chapter 23: Testosterone use in women

estrogens in ovarian and peripheral States: prevalence and predictors. hypoactive sexual desire. Fertil Steril
venous blood. J Clin Endocrinol 39: Jama 281:531544 79:13411352
10201024 Longcope C (1986) Adrenal and
Lawson EA, Miller KK, Bredella MA,
Judd HL, Lucas WE, Yen SSC (1994) Phan C, Misra M, Meenaghan E, gonadal androgen secretion in
Effect of oopherectomy on Rosenblum L, Donoho D, Gupta R, normal females. J Clin Endocrinol
circulating testosterone and Klibanski A (2010) Hormone 15:213228
androstenedione levels in patients predictors of abnormal bone Loukovaara M, Carson M,
with endometrial cancer microarchitecture in women with Adlercreutz H (1995) Regulation of
118:793798 anorexia nervosa. Bone 46:458463 production and secretion of sex
Kalantaridou SN, Calis KA, Leao LM, Duarte MP, Silva DM, Bahia hormone-binding globulin in
Vanderhoof VH, Bakalov VK, PR, Coeli CM, de Farias ML (2006) HepG2 cell cultures by hormones
Corrigan EC, Troendle JF, Influence of methyltestosterone and growth factors. J Clin
Nelson LM (2006) Testosterone postmenopausal therapy on plasma Endocrinol Metab 80:160164
deficiency in young women with 46, lipids, inflammatory factors, glucose Manderson L (2005) The social and
XX spontaneous premature ovarian metabolism and visceral fat: a cultural context of sexual function
failure. Fertil Steril 86:14751482 randomized study. Eur J Endocrinol among middle-aged women.
154:131139 Menopause 12:361362
Kasperk CH, Wergedal JE, Farley JR,
Llinkhart TA, Turner RT, Lee JS, LaCroix AZ, Wu L, Cauley JA, Mathur RS, Landgreve SC, Moody LO,
Baylink DG (1989) Androgens Jackson RD, Kooperberg C, Semmens JP, Williamson HO
directly stimulate proliferation of Leboff MS, Robbins J, Lewis CE, (1985) The effect of estrogen
bone cells in vitro. Endocrinology Bauer DC, Cummings SR (2008) treatment on plasma concentrations
124:15761578 Associations of serum sex of steroid hormones,
hormone-binding globulin and sex gonadotropins, prolactin and sex
Kocoska-Maras L, Zethraeus N,
hormone concentrations with hip hormone-binding globulin in
Radestad AF, Ellingsen T, von
fracture risk in postmenopausal post-menopausal women. Maturitas
Schoultz B, Johannesson M,
Hirschberg AL (2011)
women. J Clin Endocrinol Metab 7:129133
93:17961803 Miller K, Sesmilo G, Schiller A,
A randomized trial of the effect of
testosterone and estrogen on verbal Leiblum S, Bachmann GA, Schonfeld D, Burton S, Klibanski A
fluency, verbal memory, and spatial Kemmann E, Colburn D, (2001) Androgen deficiency in
ability in healthy postmenopausal Schwartzman L (1983) The women with hypopituitarism. J Clin
women. Fertil Steril 95:152157 importance of sexual activity and Endocrinol Metab 86:561567
hormones. Jama 249:2195 Miller KK, Biller BM, Beauregard C,
Krug R, Psych D, Pietrowsky R, Fehm
HL, Born J (1994) Selective Lellamo F, Volterrani M, Caminiti G, Lipman JG, Jones J, Schoenfeld D,
influence of menstrual cycle on Karam R, Massaro R, Fini M, Sherman JC, Swearingen B,
perception of stimuli with Collins P, Rosano GM (2010) Loeffler J, Klibanski A (2006) Effects
reproductive significance. Testosterone therapy in women of testosterone replacement in
Psychosom Med 56:410417 with chronic heart failure: a pilot androgen-deficient women with
double-blind, randomized, placebo- hypopituitarism: a randomized,
Labrie F (1991) Intracrinology. Mol
controlled study. J Am Coll Cardiol double-blind, placebo-controlled
Cell Endocrinol 78:C113C118
56:13101316 study. J Clin Endocrinol Metab
Labrie F, Martel C, Balser J (2010) Lindemann K, Vatten LJ, Ellstrom- 91:16831690
Wide distribution of the serum Engh M, Eskild A (2008) Body Miller KK, Lawson EA, Mathur V,
dehydroepiandrosterone and sex mass, diabetes and smoking, Wexler TL, Meenaghan E, Misra M,
steroid levels in postmenopausal and endometrial cancer risk: a Herzog DB, Klibanski A (2007)
women: role of the ovary? follow-up study. Br J Cancer Androgens in women with anorexia
Menopause 18:3043 98:15821585 nervosa and normal-weight women
Laughlin G, Barrett-Connor E, Kritz- Lobo R, Rosen RC, Yang H-M, with hypothalamic amenorrhea.
Silverstein D, Von Muhlen D (2000) Block B, Van der Hoop R (2003) J Clin Endocrinol Metab 92:
Hysterectomy, oophorectomy, and Comparative effects of oral 13341339
endogenous sex hormone levels in esterified estrogens with and Moller MC, Bartfai AB, Radestad AF
older women: the Rancho Bernado without methyl testosterone on (2010) Effects of testosterone
Study 85:645 endocrine profiles and dimensions and estrogen replacement on
Laumann E, Paik A, Rosen RC (1999) of sexual function in memory function. Menopause
Sexual dysfunction in the United postmenopausal women with 17:983989

513
Chapter 23: Testosterone use in women

Montalcini T, Gorgone G, Perrone AM, Cerpolini S, levels of sex steroid hormones in


Gazzaruso C, Sesti G, Perticone F, Maria Salfi NC, Ceccarelli C, men and women during
Pujia A (2007) Endogenous De Giorgi LB, Formelli G, normal aging and in Alzheimers
testosterone and endothelial Casadio P, Ghi T, Pelusi G, Pelusi C, disease. Neurobiol Aging 32:604613
function in postmenopausal Meriggiola MC (2009) Effect Sarrel P, Dobay B, Wiita B (1998)
women. Coron Artery Dis 18:913 of long-term testosterone Estrogen and estrogen-androgen
Mushayandebvu T, Castracane DV, administration on the endometrium replacement in postmenopausal
Gimpel T, Adel T, Santoro N (1996) of female-to-male (FtM) women dissatisfied with estrogen-
Evidence for diminished midcycle transsexuals. J Sex Med 6:31933200 only therapy. Sexual behavior and
ovarian androgen production in Pike CJ, Carroll JC, Rosario ER, neuroendocrine responses. J Reprod
older reproductive aged women. Barron AM (2009) Protective Med 43:847856
Fertil Steril 65:721723 actions of sex steroid hormones Selva DM, Hammond GL (2009)
Naessen T, Sjogren U, Bergquist J, in Alzheimers disease. Front Peroxisome-proliferator receptor
Larsson M, Lind L, Kushnir MM Neuroendocrinol 30:239258 gamma represses hepatic sex
(2010) Endogenous steroids Procter and Gamble Pharmaceuticals hormone-binding globulin
measured by high-specificity liquid (2004) Intrinsa (testosterone expression. Endocrinology
chromatography-tandem mass transdermal system) NDA No. 21 150:21832189
spectrometry and prevalent 769. Ed ACfRH Drugs. US Food Selva DM, Hogeveen KN, Innis SM,
cardiovascular disease in 70-year- and Drug Administration, Hammond GL (2007)
old men and women. J Clin Washington, DC Monosaccharide-induced
Endocrinol Metab 95:18891897 Pugeat M, Cousin P, Baret C, Lejeune H, lipogenesis regulates the human
Ness RB, Albano J, McTiernan A, Forest MG (2000) Sex hormone- hepatic sex hormone-binding
Cauley J (2009) Influence of binding globulin during puberty in globulin gene. J Clin Invest
estrogen plus testosterone normal and hyperandrogenic girls. 117:39793987
supplementation on breast cancer. J Pediatr Endocrinol Metab Shah SM, Bell RJ, Savage G, Goldstat R,
Arch Intern Med 169:4146 13 (Suppl 5):12771279 Papalia MA, Kulkarni J, Donath S,
Nestler JE, Strauss JF 3rd (1991) Randolph JF Jr, Sowers M, Gold EB, Davis SR (2006) Testosterone
Insulin as an effector of human Mohr BA, Luborsky J, Santoro N, aromatization and cognition in
ovarian and adrenal steroid McConnell DS, Finkelstein JS, women: a randomized, placebo-
metabolism. Endocrinol Metab Clin Korenman SG, Matthews KA, controlled trial. Menopause
North Am 20:807823 Sternfeld B, Lasley BL (2003) 13:600608
Nurnberg HG, Hensley PL, Heiman JR, Reproductive hormones in the early Sherwin BB, Gelfand MM (1987) The
Croft HA, Debattista C, Paine S menopausal transition: relationship role of androgen in the maintenance
(2008) Sildenafil treatment of to ethnicity, body size, and of sexual functioning in
women with antidepressant- menopausal status. J Clin Endocrinol oophorectomized women.
associated sexual dysfunction: a Metab 88:15161522 Psychosomatic Med 49:397409
randomized controlled trial. Jama Rariy CM, Ratcliffe SJ, Weinstein R, Shifren JL, Braunstein G, Simon J,
300:395404 Bhasin S, Blackman MR, Cauley JA, Casson P, Buster JE, Red Burki RE,
Ouyang P, Vaidya D, Dobs A, Robbins J, Zmuda JM, Harris TB, Ginsburg ES, Rosen RC, Leiblum
Golden SH, Szklo M, Heckbert SR, Cappola AR (2011) Higher serum SR, Caramelli KE (2000)
Kopp P, Gapstur SM (2009) Sex free testosterone concentration in Transdermal testosterone treatment
hormone levels and subclinical older women is associated with in women with impaired sexual
atherosclerosis in postmenopausal greater bone mineral density, lean function after oophorectomy. N Eng
women: the Multi-Ethnic Study of body mass, and total fat mass: the J Med 343:682688
Atherosclerosis. Atherosclerosis cardiovascular health study. J Clin
Endocrinol Metab 96:989996 Shifren JL, Davis SR, Moreau M,
204:255261 Waldbaum A, Bouchard C,
Panay N, Al-Azzawi F, Bouchard C, Rieche K, Wolff G (1975) Comparison DeRogatis L, Derzko C, Bearnson C,
Davis SR, Eden J, Lodhi I, Rees M, of testosterone decanoate, Kakos N, ONeill S, Levine S,
Rodenberg CA, Rymer J, drostanolone and testololactone in Wekselman K, Buch A,
Schwenkhagen A, Sturdee DW disseminated breast cancer a Rodenberg C, Kroll R, Kroll M
(2010) Testosterone treatment of randomized clinical study. Arch (2006) Testosterone patch for the
HSDD in naturally menopausal Geschwulstforsch 45:485488 treatment of hypoactive sexual
women: the ADORE study. Rosario ER, Chang L, Head EH, desire disorder in naturally
Climacteric 13:121131 Stanczyk FZ, Pike CJ (2009) Brain menopausal women: results from

514
Chapter 23: Testosterone use in women

the INTIMATE NM1 study. temperature. J Steroid Biochem sexual arousal in women. Arch Gen
Menopause 13:770779 16:801810 Psychiatry 57:149
Shifren JL, Monz BU, Russo PA, Somboonporn W, Davis S (2004) Tungphaisal S, Chandeying V,
Segreti A, Johannes CB (2008) Testosterone and the breast: Sutthijumroon S, Krisanapan O,
Sexual problems and distress in therapeutic implications for Udomratn P (1991) Postmenopausal
United States women: prevalence women. Endocrine Reviews sexuality in Thai women. Asia
and correlates. Obstet Gynecol 25:374388 Oceania J Obstet Gynaecol 17:
112:970978 143146
Somboonporn W, Bell RJ, Davis SR
Sievers C, Klotsche J, Pieper L, (2005) Testosterone for peri and Utian WH (1972) The true clinical
Schneider HJ, Marz W, postmenopausal women. features of postmenopausal
Wittchen HU, Stalla GK, Mantzoros Cochrane Database Syst Rev 4: oophorectomy and their response to
C (2010) Low testosterone levels CD004509 estrogen replacement therapy. S Afr
predict all-cause mortality and Med J 46:732737
Sutton-Tyrrell K, Wildman RP,
cardiovascular events in women: a Wagner JD, Zhang L, Williams JK,
Matthews KA, Chae C, Lasley BL,
prospective cohort study in German Register TC, Ackerman DM,
Brockwell S, Pasternak RC, Lloyd-
primary care patients. Eur Wiita B, Clarkson TB, Adams MR
Jones D, Sowers MF, Torrens JI
J Endocrinol 163:699708 (1996) Esterified estrogens with
(2005) Sex hormone-binding
Simon J, Braunstein G, Nachtigall L, globulin and the free androgen and without methyltesterone
Utian W, Katz M, Miller S, index are related to cardiovascular decrease arterial LDL metabolism
Waldbaum A, Bouchard C, risk factors in multiethnic in Cynomolgus monekys.
Derzko C, Buch A, Rodenberg C, premenopausal and perimenopausal Arterioscler Thromb Vasc Biol
Lucas J, Davis S (2005) Testosterone women enrolled in the Study of 16:14731480
patch increases sexual activity and Women Across the Nation Warnock JK, Swanson SG, Borel RW,
desire in surgically menopausal (SWAN). Circulation Zipfel LM, Brennan JJ (2005)
women with hypoactive sexual 111:12421249 Combined esterified estrogens and
desire disorder. J Clin Endocrinol methyltestosterone versus esterified
Metab 90:52265233 Taieb J, Mathian B, Millot F, Patricot
MC, Mathieu E, Queyrel N, estrogens alone in the treatment of
Simpson ER, Davis SR (2001) Lacroix I, Somma-Delpero C, loss of sexual interest in surgically
Minireview: aromatase and the Boudou P (2003) Testosterone menopausal women. Menopause
regulation of estrogen biosynthesis measured by 10 immunoassays 12:374384
some new perspectives. and by isotope-dilution gas Watts NB, Notelovitz M,
Endocrinology 142:45894594 chromatography-mass spectrometry Timmons MC (1995) Comparison
Slemenda C, Longcope C, Peacock M, in sera from 116 men, women, and of oral estrogens and estrogens plus
Hui S, Johnston CC (1996) Sex children. Clin Chem 49:13811395 androgen on bone mineral density,
steroids, bone mass, and bone loss. Tamimi RM, Hankinson SE, Chen WY, menopausal symptoms and lipid-
A prospective study of pre-, peri- Rosner B, Colditz GA (2006) lipoprotein profiles in surgical
and postmenopausal women. J Clin Combined estrogen and menopause 85:529537
Invest 97:1421 testosterone use and risk of breast Werneke U, Northey S, Bhugra D
Sluijmer AV, Heineman MJ, cancer in postmenopausal women. (2006) Antidepressants and sexual
De Jong FH, Evers JL (1995) Arch Intern Med 166:14831489 dysfunction. Acta Psychiatr Scand
Endocrine activity of the Thurlimann B, Keshaviah A, 114:384397
postmenopausal ovary: the effects of Coates AS, Mouridsen H, Mauriac Wild S, Pierpoint T, Jacobs H,
pituitary down-regulation and L, Forbes JF, Paridaens R, McKeigue P (2000) Long-term
oophorectomy. J Clin Endocrinol Castiglione-Gertsch M, Gelber RD, consequences of polycystic ovary
Metab 80:21632167 Rabaglio M, Smith I, Wardly A, syndrome: results of a 31 year
Snabes MC, Simes SM (2009) Price KN, Goldhirsch A (2005) follow-up study. Hum Fertil (Camb)
Approved hormonal treatments for A comparison of letrozole and 3:101105
HSDD: an unmet medical need. tamoxifen in postmenopausal Wisniewski A, Ngyuen T, Dobs AS
J Sex Med 6:18461849 women with early breast cancer. (2002) Evaluation of high-dose
Sodergard R, Backstrom T, Shanhag V, N Engl J Med 353:27472757 estrogen and high-dose estrogen
Carstensen H (1982) Calculation of Tuiten A, Von Honk J, Koppeschaar H, plus methyltestosterone treatment
free and bound fractions of Bernaards C, Thijssen J, Verbaten R on cognitive task performance in
testosterone and estradiol-17 beta to (2000) Time course of effects of postmenopausal women. Horm Res
human plasma proteins at body testosterone administration on 58:150155

515
Chapter 23: Testosterone use in women

Witherby S, Johnson J, Demers L, receiving estrogen. J Clin Endocrinol Zang H, Sahlin L, Masironi B,
Mount S, Littenberg B, Metab 86:158161 Eriksson E, Linden Hirschberg A
Maclean CD, Wood M, Muss H Yao M, Nguyen TV, Rosario ER, (2007) Effects of testosterone
(2011) Topical testosterone for Ramsden M, Pike CJ (2008) treatment on endometrial
breast cancer patients with vaginal Androgens regulate neprilysin proliferation in postmenopausal
atrophy related to aromatase expression: role in reducing beta- women. J Clin Endocrinol Metab
inhibitors: a phase I/II study. amyloid levels. J Neurochem 92:21692175
Oncologist 16:424431 105:24772488 Zuckerman-Levin N, Frolova-Bishara T,
Wolf OT, Kirschbaum C (2002) Zang H, Davis SR (2008) Androgen Militianu D, Levin M, Aharon-
Endogenous estradiol and replacement therapy in androgen- Peretz J, Hochberg Z (2009)
testosterone levels are associated deficient women with Androgen replacement therapy in
with cognitive performance in older hypopituitarism. Drugs Turner syndrome: a pilot study.
women and men 41:259 68:20852093 J Clin Endocrinol Metab
Worboys S, Kotsopoulos D, Teede H, Zang H, Carlstrom K, Arner P, 94:48204827
McGrath BP, Davis SR (2001) Hirschberg AL (2006) Effects of Zumoff B, Strain GW, Miller LK,
Evidence that parenteral treatment with testosterone alone or Rosner W (1995) Twenty-four hour
testosterone therapy may improve in combination with estrogen on mean plasma testosterone
endothelium-dependent insulin sensitivity in concentration declines with age in
and -independent vasodilation in postmenopausal women. Fertil Steril normal premenopausal women
postmenopausal women already 86:136144 80:14291430

516
Chapter
Detection of illegal use of androgens and

24 selective androgen receptor modulators


Wilhelm Schanzer and Mario Thevis

24.1 Introduction 517 24.6.2.4 Derivatization 526


24.2 Frequency of steroid hormone misuse 520 24.6.3 Detection of synthetic anabolic
24.2.1 Androgen misuse in controlled androgenic steroids 526
competition sports 520 24.6.4 Detection of endogenous anabolic
24.2.2 Androgen misuse in non-competitive androgenic steroids 528
sports 520 24.6.4.1 Indirect detection methods 528
24.3 Prohormones of androgens 521 24.6.4.2 Direct detection method: gas
chromatographycombustion
24.4 Designer steroids 522
isotope ratio mass spectrometry
24.5 Contamination of nutritional supplements (GC-C-IRMS) 528
with prohormones 523
24.7 Selective androgen receptor modulators
24.6 Detection of misuse of anabolic androgenic (SARMs) 529
steroid hormones 523
24.7.1 General aspects 529
24.6.1 Organization of doping tests 523
24.7.2 Analytical approaches 529
24.6.2 Detection and identification of misused
24.8 Outlook 530
anabolic androgenic steroids 524
24.9 Key messages 531
24.6.2.1 Metabolism 525
24.10 References 531
24.6.2.2 Pharmacokinetics 525
24.6.2.3 Sample preparation 525

24.1 Introduction By using these steroids, athletes hoped to increase


Anabolic androgenic steroids (AAS) are known to muscle strength. Such improvements in muscle
be misused both in competitive and in non-com- strength to increase physical performance in sport
petitive sports (Haupt and Rovere 1984; Wilson are naturally an essential effect of training. As AAS
1988; Yesalis et al. 1993). Moreover, it seems that stimulate protein synthesis in muscle cells, athletes
AAS are becoming social drugs, as even young expect performance-enhancing effects beyond those
people apply them as an expression of an improved brought about by training alone. At the end of the
lifestyle. sixties, the first anti-doping rules were established by
The misuse of AAS in athletics has been observed international sport federations (1967, International
for more than 50 years. The first rumors dated from Cycling Union and 1968, International Olympic
1954 and were attributed to weightlifters who seemed Committee IOC), but only stimulants and narcotics
to have used testosterone (Wade 1972). By 1965 syn- were banned (Clasing 1992). At that time, the Medical
thetic AAS had become widely popular among body- Commission of the IOC was already aware of wide-
builders and weightlifters, but were also applied in spread misuse of AAS in sports. They were not
other forms of sports. banned because no reliable method was available to

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

517
Chapter 24: Detecting illegal use of androgens and SARMs

detect them (Beckett and Cowan 1979). Under these developed but misused to a lesser extent, e.g. 11b-
circumstances the first methods for AAS detection hydroxy-Oral Turinabol, also named as SXII. Con-
were developed (Brooks et al. 1975; Ward et al. cerning performance-enhancement effects, Franke
1975), and in 1974 the Medical Commission of the and Berendonk (1997) quoted a summary report
IOC and the International Amateur Athletic Feder- to the Stasi on March 3, 1977 . . . (Stasi = Ministry
ation (IAAF) first banned the use of AAS. This pro- of State Security) under the code name Technik,
hibition encompassed only synthetic steroids, such as (pp. 24344)), which described the GDR results
metandienone, stanozolol etc., and the use of and concluded:
endogenous steroids, e.g. testosterone, was not
The positive value of anabolic steroids for the
restricted. At that time athletes misused only synthetic
development of a top performance is undoubted. Here
AAS. The reason was that AAS were used in human are a few examples . . . Performances could be improved
medicine to treat catabolic conditions; scientific data with the support of these drugs within four years as
obtained from animal studies led to the conclusion follows: Shot-put (men) 2.54 m; Shot-put (women)
that synthetic AAS are more anabolic and less andro- 4.55 m; Discus throw (men) 1012 m; Discus throw
genic than testosterone itself (Kochakian 1976). (women) 1120 m; Hammer throw 610 m; Javelin
Whether athletes experience a positive performance- throw (women) 815 m; 400 m (women) 45 sec; 800
enhancing effect or not when using AAS has been m (women) 510 sec; 1500 m (women) 710 sec . . .
discussed controversially for many years. Nowadays Remarkable rates of increase in performances were also
it is known that androgens have muscle growth- noted in the swimming events of women . . . from our
promoting effects in boys, in women and in hypo- experiences made so far, it can be concluded that
women have the greatest advantage from treatments
gonadal men. It has never been proven that andro-
with anabolic hormones with respect to their
gens, when administered in therapeutic doses, have performance in sports . . . Especially high is the
positive effects on muscle growth in healthy adult performance-supporting effect following the first
men. The assumption that AAS have less effect on administration of anabolic hormones, especially with
muscle growth in men is based on the fact that the junior athletes.
androgen receptor in men is nearly completely sat-
urated (Wilson 1996). However, high doses of To control the (mis)use of synthetic AAS, urine
androgens have been reported to exert muscle samples of athletes collected after competition events
mass-enhancing effects (Bhasin et al. 1996). An were tested. As AAS are not used directly during
unethical and secret program of hormonal doping competition but rather during training to increase
of athletes in the former German Democratic muscle strength, athletes stopped administration of
Republic (GDR) was reported (Franke and Beren- AAS before competition, switching to those AAS
donk 1997), and performance-improving effects of (e.g. stanozolol) they believed could not be detected,
AAS were elucidated. and to endogenous androgens, such as testosterone,
The most frequently misused steroid in the which were not banned. Investigations of test samples
GDR was Oral Turinabol (dehydrochloromethyl- from the Olympic Summer Games in 1980 in
testosterone, Fig. 24.1), which was used from 1968 Moscow showed that 2.1% of male and 7.1% of female
onwards by nearly all male and female athletes. In athletes had elevated testosterone levels (Zimmer-
later years the steroid mestanolone (the 17a-meth- mann 1986) in urine, and the highest urinary testos-
ylated dihydrotestosterone, 17b-hydroxy-17a- terone levels were detected for women in swimming
methyl-5a-androstan-3-one) was also regularly and track and field events. These results could only be
administered, which seemed to be more effective explained by exogenous application of testosterone.
in promoting aggressiveness and allowed higher Donike developed a GC-MS method for detection
training cycles without much bodyweight increase of testosterone and epitestosterone excreted in urine,
(Franke and Berendonk 1997). Beside oral andro- and proved that the ratio of testosterone to epitestos-
genic steroids, also injections of steroid esters (nor- terone (T/E) is significantly increased after applica-
testosterone and testosterone esters) were applied, tion of testosterone (Donike et al. 1983). Based on
mainly to female athletes. Interestingly, in the these results, sport federations also banned testoster-
document of Franke and Berendonk (1997), further one in 1984 and applied the T/E ratio measurements
chlorinated steroids are listed which have been (cutoff level of six) to their rules. In 2006 the World

518
Chapter 24: Detecting illegal use of androgens and SARMs

OH OH Fig. 24.1 Structural formulae of AAS


OH which have been internationally detected
in doping tests in sports.

O O
O H
Cl
Boldenone Clostebol Dihydrotestosterone

OH OH OH
CH3 HO CH3
H3C
F
O O O
Cl H
Dehydrochloromethyl- Drostanolone Fluoxymesterone
testosterone
OH OH
OH
CH3
HO CH3 CH3
O
H C

O O
O H
Formebolone Mesterolone Metandienone

OH
OH OH
CH3 CH3
CH3

O H O O
Metenolone Methyltestosterone Methyltrienolone
OH OH
OH
CH3 CH3
O
HC
H N
O N
H
O H
Nortestosterone Oxymetholone Stanozolol

OH OH OH
CH3

O O O
Oxandrolone Testosterone Trenbolone

Anti-Doping Agency (WADA) reduced the T/E ratio such as dihydrotestosterone, and prohormones of
cutoff to four, and athletes with increased values will testosterone, dihydrotestosterone and nortestoster-
undergo follow-up investigations. In addition to the one, as well as new designer steroids, were added to
ban of testosterone, other endogenous androgens the list of prohibited AAS during recent years.

519
Chapter 24: Detecting illegal use of androgens and SARMs

24.2 Frequency of steroid worldwide in comparison to 1094 cases for stanozolol


and 526 for metandienone.
hormone misuse
Considering this misuse of AAS, the question arises,
to what extent are AAS really misused? Is it restricted
24.2.2 Androgen misuse in non-
to a few high-level performing athletes in sports or is competitive sports
it so extensive as to be regarded as a social drug In comparison to officially controlled competition
problem? Available data derive: sports, no analytical data from laboratories concern-
1. from positive findings resulting from checking ing the misuse of AAS are available for those areas of
AAS doping in competition sports; and athletics and private life where no tests are performed.
2. from results of questionnaires concerning the use To overcome this lack of information, scientists have
of AAS in non-competitive sports. performed surveys; however, only a few publications
are available. Yesalis et al. (1993) published results of
investigations in the United States and calculated that
24.2.1 Androgen misuse in controlled one million Americans had used AAS some time in
their lives, including about 250 000 in the past year. In
competition sports 1993 a Canadian study (Canadian Centre for Drug-
Androgen misuse in competition sports is investi- Free Sport 1993) confirmed that, in Canada, 80 000
gated by laboratories which are accredited by the young people between the ages of 13 and 18 had used
WADA. Each year the accredited laboratories (35 AAS. A self-report questionnaire about the misuse of
laboratories in 2011, worldwide) report the positive AAS among 13 355 Australian high-school students
findings from the A-sample (see below) analyses. The reported 3.2% of male and 1.2% of female users
annual testing frequency was about 207 500 samples (Handelsman and Gupta 1997). Questionnaires from
for all laboratories from 2002 to 2010. In this time Switzerland were summarized by Kamber (1995),
period a total number of 1 867 109 doping tests were who concluded that AAS are a serious problem.
analyzed by the WADA-accredited laboratories for A questionnaire from 16 000 recruits in Switzerland
Olympic and non-Olympic sports, and 9345 AAS and 3700 women of the same age showed that 1.8% of
were reported in different types of sport as positive the recruits and 0.3% of the women had administered
findings. This represents 0.5% of all test samples. AAS in 1993. Two surveys in Germany investigated
Compared to data from 19922001, the positive find- the misuse of AAS in fitness studios mainly associated
ings with AAS were 0.34% lower. Fig. 24.1 shows the with bodybuilding. Boos et al. (1998) published a
chemical structure of misused steroids. The data indi- misuse rate of AAS of about 24% in males and 8%
cate that the misuse of AAS is limited to a number of in females, and Striegel et al. (2006) reported a rate of
well-known AAS. Among the most frequent steroid 19.2% in male and 3.9% in female visitors in fitness
hormones misused in controlled competitive sports centers.
are the synthetic steroids nortestosterone, stanozolol, Data concerning the most commonly misused
metandienone and metenolone, and the endogenous AAS in non-controlled sports are only available via
steroid testosterone. recommendations in magazines for bodybuilders, via
The detection of Oral Turinabol, the most widely underground handbooks (Taylor 1982; Duchaine
used steroid in the former GDR, is based on identifi- 1989; Grundig and Bachmann 1995; Sinner 2009)
cation of long-term excreted metabolites following and the internet and via confiscated, smuggled sub-
hydroxylation at C-6, C-16, 17-epimerization and stances and those obtained from black-market
reduction of the 4,5 double bond to a 5b-configur- sources. Frequently recommended AAS include
ation. The steroid is still of interest and in recent years boldenone undecylenate, drostanolone enanthate
it appeared on the black market, advertised as a new and propionate, fluoxymesterone, mesterolone,
reinvented steroid. But the numbers of positive metandienone, metenolone acetate and enanthate,
results in doping control are still low compared to methandriol dipropionate, methyltestosterone, nor-
the misuse of metandienone and stanozolol. In the testosterone decanoate and other esters (hexylphenyl-
time period from 2005 to 2009, approximately 29 propionate, laurate, propionate and undecanoate),
doping violations were reported for Oral Turinabol oxandrolone, oxymetholone, stanozolol, testosterone

520
Chapter 24: Detecting illegal use of androgens and SARMs

O O OH Fig. 24.2 Structural formulae of


prohormones of testosterone,
dihydrotestosterone and 1-ene steroids.

HO O HO
Dehydroepiandrosterone 4-Androstenedione 4-Androstenediol
OH OH
OH

HO HO
HO H H
5-Androstenediol 5-Androstane-3,17-diol 5-Androstane-3,17-diol

O O OH

O O O
H H H
5-Androstanedione 1-Androstenedione 1-Testosterone

OH
O OH

HO
H O HO
1-Androstenediol 4-Norandrostendione 4-Norandrostenediol

in the form of different esters (cypionate, enanthate, structure (1-androstenedione (5a-androst-1-ene-


heptylate, decanoate, hexanoate, isocaproate, isohex- 3,17-dione), 1-testosterone (17b-hydroxy-5a-
anoate, phenylpropionate, propionate and undecano- androst-4-en-3-one), 1-androstenediol (5a-androst-
ate) and trenbolone, trenbolone acetate. 1-ene-3b,17b-diol)) (Fig. 24.2), and prohormones of
These substances are largely identical with those nortestosterone (4-norandrostenedione (estr-4-ene-
products which were confiscated and distributed by 3,17-dione), 4-norandrostenediol (estr-4-ene-3b,17b-
the black market over the last 20 years. diol)) (Fig. 24.2) have been marketed in the United
States as nutritional supplements.
Steroids with 5a-androst-1-ene structure may be
24.3 Prohormones of androgens considered as prohormones of dihydrotestosterone,
Since 1999, prohormones of testosterone (e.g. dehy- as the double bond at C12 is partly hydrogenated
droepiandrosterone (DHEA, 3b-hydroxyandrost-5- in human metabolism, but otherwise the compounds
en-17-one), 4-androstenedione (androst-4-ene-3,17- are not synthesized in the human body as intermedi-
dione), 4-androstenediol (androst-4-ene-3b,17b- ates of endogenously produced steroids and should be
diol), 5-androstenediol (androst-5-ene-3b,17b-diol)) classified as designer steroids.
(Fig. 24.2), prohormones of dihydrotestosterone Prohormones are advertised as having effects
(5a-androstane-3b,17b-diol, 5a-androstane-3a,17b- similar to testosterone, dihydrotestosterone and nor-
diol, 5a-androstanedione (5a-androstane-3,17- testosterone because of a high conversion rate of
dione)) (Fig. 24.2), steroids with 5a-androst-1-ene prohormones to the physiologically effective steroids

521
Chapter 24: Detecting illegal use of androgens and SARMs

in the human body after oral, sublingual or buccal of controlled substances by the Anabolic Steroid
application. In contrast to such incorrect advertise- Control Act to prohibit the illegal distribution of
ments, only small amounts of the applied prohor- these substances. In Europe prohormones are con-
mone may be converted to the effective steroid. sidered unlicensed medications, and their distribution
Indeed, for medical treatment prohormones are use- is banned. Nevertheless, control of the misuse of
less. Therefore companies providing prohormones prohormones is difficult: products enter the European
recommend applying several times per day, especially market via neighbor states, directly via airports and
before training or competition (high amounts (100 by internet or postal orders.
mg and more) of oral preparations of single prohor- Additional new products have entered the market
mones, combinations of different prohormones and with a 1-ene structure, such as androsta-1,4-diene-
sublingual preparations). Published data (Leder et al. 3,17-dione, which is considered as a prohormone of
2000) demonstrate that, in male persons, e.g. andros- the synthetic AAS boldenone (17b-hydroxyandrosta-
tenedione 100 mg/day taken orally for seven days 1,4-dien-3-one), and steroids with a 5a-androst-1-ene
yielded no significant changes in serum testosterone structure (Fig. 24.2) which are marketed as
levels compared to the control group, whereas prohormones.
300 mg/day of androstenedione for seven days
showed a significant increase in peak and AUC (area
under curve) serum testosterone (AUC 34%), but 24.4 Designer steroids
with high interindividual variation. As prohormones Designer steroids are anabolic steroids which are
are also used by females and adolescents, lower designed to be undetectable in drug testing but have
amounts of prohormones may yield physiological anabolic effects similar to testosterone and other ster-
changes in serum testosterone levels; e.g. 100 mg oid-like performance-enhancing drugs. The most
androstenedione given orally to females yielded an famous scandal with designer steroids was the Balco
increase in serum testosterone concentrations of up scandal in 2002 when the sports nutrition center, Bay
to 0.8 ng/ml (Kicman et al. 2003). Area Laboratory Co-operative (BALCO), in Califor-
In a study with 19-norandrostenediol, a prohor- nia (USA) provided a modified steroid to top-level
mone of 19-nortestosterone, by Schrader et al. (2006), athletes. This steroid was later identified as tetrahy-
an open-label, crossover trial with eight healthy male drogestrinone (THG). The starting steroid was an
volunteers was conducted. After administration of already-banned anabolic androgenic steroid named
capsules or sublingual tablets with 25 mg of 19-nor- gestrinone, which was chemically modified by the
androstenediol, plasma concentrations of the introduction of four hydrogen atoms at the C-17
pharmacologically active 19-nortestosterone were ethinyl group (Fig. 24.3). The tetrahydrogestrinone
determined up to 5.7 ng/ml. The results demonstrate so obtained remained undetectable until reference
the importance of prohibiting prohormones such as material was delivered to the IOC-accredited doping
19-norandrostenediol, in particular, since plasma control laboratory in Los Angeles (Catlin et al. 2004).
concentrations of nandrolone between 0.3 and 1.2 Further designer steroids such as norboletone (Catlin
ng/ml have been reported to influence endocrino- et al. 2002) and desoxymethyltestosterone (Madol,
logical parameters (Belkien et al. 1985). DMT) (Sekera et al. 2005) were distributed at the
The distribution of prohormones in the United same time. Since that time, an increasing number of
States was not restricted until 2005 because these designer steroids, not explicitly synthesized for ath-
products have not been marketed as medications. letes, have also been illegally marketed and advertised
But in 2005 prohormones were included in the list as dietary supplements (Kazlauskas 2010; Parr and

OH Fig. 24.3 Structural formulae of


H5C2 OH H5C2 OH
CH3 designer steroids Madol, norboletone and
C2H5 CH2CH3 tetrahydrogestrinone.
H

H
H O O
Desoxymethyltestosterone Norboletone Tetrahydrogestrinone (THG)
(Madol)

522
Chapter 24: Detecting illegal use of androgens and SARMs

Schnzer 2010). None of these designer steroids is of supplements offered in western countries and which
interest for medical purposes. Depending on the have been identified as a source for positive findings.
country, they are illegally distributed. They are non- The problem was clearly in connection with prohor-
approved drugs and may cause severe side-effects. mones of testosterone and nortestosterone produced
in the United States and was not aimed at products
24.5 Contamination of nutritional originating from Asian countries.)
The positive supplements showed anabolic andro-
supplements with prohormones genic steroid concentrations of 0.01 mg/g up to 190
Since 1999, the same time when prohormones were mg/g. Excretion studies with application of supple-
entering the market, nutritional supplements have ments containing nortestosterone prohormones cor-
become sporadically contaminated with prohor- responding to a total uptake of more than 1 mg
mones. Contamination in this context signifies that resulted in urinary concentrations of the nortestoster-
a supplement contains substances which are not one metabolite norandrosterone above the cutoff
declared on the label. As the amount of contamin- limit (2 ng/ml urine) for several hours (positive
ation is low, less than 1 per mille of the product, it doping result).
is assumed that the prohormones are not intention- Positive doping tests caused by contaminated
ally added to the supplements but may be contamin- supplements with prohormones of only testosterone
ated due to poor quality control during the or dihydrotestosterone have not been proved, which
production of nutritional supplements and prohor- is explainable by the low amounts of contamination
mones. Several athletes have been tested in the past and the applied tests, which have to differentiate
with positive results for norandrosterone (main between endogenous and exogenous origin of testos-
metabolite of nortestosterone and the main metabol- terone and which will not be influenced by ingestion
ite of the prohormones of nortestosterone such as of low amounts of prohormones or testosterone itself.
norandrost-4-ene-3,17-dione and norandrost-4-ene- This problem of contaminated nutritional supple-
3b,17b-diol); the use of nutritional supplements ments with prohormones seems to have beeen
which contained low traces of prohormones of nor- reduced in recent years, as in 2005 the Anabolic
testosterone led to positive results. In such a case the Steroid Control Act in the United States banned
athlete has not intentionally applied a doping sub- the marketing and distribution of prohormones.
stance, but the sports federations consider the pres-
ence of norandrosterone in the urine sample as a
doping offence for which the athlete is fully 24.6 Detection of misuse of anabolic
responsible. androgenic steroid hormones
To what extent nutritional supplements may be
contaminated has been shown in different studies and 24.6.1 Organization of doping tests
by an international study supported by the IOC Doping control is organized by national and inter-
(Geyer et al. 2004). The latter study investigated 630 national sports federations and by the WADA for the
different nutritional supplements from 13 countries different types of sports. Increasingly, national anti-
including the United States, Italy, France, Germany doping programs are managing doping control via
and Great Britain. Out of the 634 samples analyzed, one overall organization. This strategy seems to be
94 (14.8%) contained prohormones not declared on the most effective testing action, as any possible inten-
the label (positive supplements). Of these 94 posi- tion by individual sports federations to hide positive
tive supplements, 23 samples (24.5%) contained pro- cases and to protect their athletes can be excluded.
hormones of nortestosterone and testosterone; 64 The IOC only performed doping tests during the
samples (68.1%) only contained prohormones of tes- Olympic winter and summer games, and it has no
tosterone; 7 samples (7.5%) only contained prohor- out of competition testing program. This deficit has
mones of nortestosterone. now been compensated for by WADA.
In relation to the total number of products pur- For a doping test, athletes are selected according
chased per country, most of the positive supplements to the rules of the responsible sports federation.
(84%) originate from companies located in the United The doping test is carried out in two steps. The
States. (The investigation was focused on nutritional first step includes the sample-taking procedure and

523
Chapter 24: Detecting illegal use of androgens and SARMs

transportation of the urine specimens to a WADA- 24.6.2 Detection and identification of


accredited laboratory. In the following step the
laboratory analyzes the sample for banned drugs. misused anabolic androgenic steroids
The sample-taking procedure is an important step. Synthetic AAS were first banned in 1974. As no
To avoid any manipulation, athletes have to deliver comprehensive analytical method for the detection
a urine sample under visual inspection by an of AAS in human urine was available at the beginning
accredited supervisor. The urine is divided into an of the seventies, new methods had to be developed.
A- and a B-sample; both samples are sealed and then The first methods were based on radioimmunoassay
transported to the laboratory. All steps during this (RIA) techniques; for instance Brooks et al. (1975)
procedure are documented and the athlete has to sign developed an antiserum for metandienone with some
a protocol of the sample-taking procedure and sealing cross-reactivity to other 17a-methyl steroids. The RIA
of samples. All handling of a urine specimen (sample- techniques were discouraging for several reasons: the
taking, transportation containers and laboratory tests) method did not consider the high degree of metabol-
must be documented and is designated as chain of ism of AAS (therefore screening for the parent steroid
custody. The laboratory is not in possession of the was less successful), the antisera had only limited
athletes name corresponding to the urine sample, and sensitivity for other steroids and the possibility of
all samples have code numbers. The reason for divid- false positives, which was not acceptable for routine
ing the urine specimen into A- and B-samples is to analysis. As early as 1975 Ward et al. presented a GC-
guarantee the best chain of custody: if the A-sample is MS method for the detection of the AAS
tested positive (the laboratory reports an adverse ana- metandienone, nortestosterone, norethandrolone
lytical finding), the B-sample can be analyzed on and stanozolol. Nevertheless, the RIA technique was
request of the athlete. For transparency the B-sample used as a screening method during the 1976 Olympic
analysis will be performed in the presence of the Games in Montreal and in Moscow in 1980, but
athlete and his advisers. If the B-analysis confirms confirmation of suspicious samples was performed
the A-result, the sample is considered as positive by GC-MS. After 1981 all IOC-accredited laboratories
and the federation can impose sanctions on the used GC-MS as the main analytical tool for AAS
athlete. identification (Donike et al. 1984; Mass et al. 1989;
Doping test samples are analyzed by WADA- Schnzer and Donike 1993).
accredited laboratories. Laboratories seeking accredit- Analysis of AAS can be divided and described in
ation have to comply with the requirements for two steps: first a sample preparation is performed
doping drug testing set by the WADA World-Anti- with the aim to separate the banned substances from
Doping Code. Additionally, the laboratory has to be the biological matrix (urine) and to reduce biological
accredited by a national accreditation body following interference (biological background). Sample prepar-
the standard of ISO 17025. The laboratory must show ation for AAS also includes a chemical modification
that it has the capability to analyze all banned sub- (derivatization) of the isolated substances to improve
stances below the specified concentration limits their analytical detectability when gas chromato-
within a controlled quality system. graphic separation techniques are applied. The second
The prerequisite for this accreditation system is a step covers the analytical measurement, which is
standardization of analytical techniques and detection based on a physical principle, mainly on gas chroma-
limits of banned substances among the different tography in combination with mass spectrometry
laboratories. Information concerning new doping (GC-MS) or tandem mass spectrometry (GC-MS/
drugs and doping techniques is rapidly distributed MS). Additionally, liquid chromatography and
in order to deal with new problems in a coordinated tandem mass spectrometry (LC-MS/MS) is used for
manner. Especially for the detection of synthetic AAS, AAS with poor gas chromatographic properties or
which are misused mainly during training periods, which are sensitive to temperature.
the laboratory has to use highly sensitive methods. The main advantage of chromatographic tech-
At the present time, 35 WADA laboratories all niques such as GC-MS, GC-MS/MS and LC-MS/MS
over the world (20 in Europe, 4 in North America, 2 is the possibility of analyzing a high number of sub-
in South America, 6 in Asia, 1 in Australia and 2 in stances within one run. This minimizes costs and
Africa) are accredited. allows a high throughput of samples. In fact it is

524
Chapter 24: Detecting illegal use of androgens and SARMs

possible to run a maximum of 50 samples per day and regarding the high number of metabolites are known
per GC-MS instrument. for the metabolism of testosterone (Kochakian 1990).

24.6.2.2 Pharmacokinetics
24.6.2.1 Metabolism A further important factor which has to be considered
To a large extent, anabolic androgenic steroids are for detection of AAS is the pharmacokinetics of the
metabolized by phase I and phase II reactions, and parent compound and its excreted metabolites. As
only a few AAS are excreted unchanged in urine for a AAS are misused during training and the number of
short period of time after administration. To detect doping tests is limited, it is desirable to detect AAS as
the misuse of AAS which are not excreted in urine or long as possible after their last administration. Analy-
only to a small extent, the analytical method cannot sis of the parent steroids and/or their metabolites,
rely on monitoring only the parent steroid but must which are excreted very rapidly, is less effective for
identify its metabolites. Detection of an AAS metab- screening analysis than the detection of metabolites
olite in urine is proof for the misuse of a banned excreted long-term: these are steroids detectable for
anabolic androgenic doping substance. This presumes the longest possible period of time after administra-
that the metabolite cannot be generated from tion (Schnzer et al. 1996; Schnzer et al. 2006). The
endogenous steroids in the body compartment. main differences between the pharmacokinetics of
Metabolites of the most frequently misused ana- AAS are caused by their pharmaceutical preparation
bolic steroids have been investigated by different and the kind of application. Depot preparations, e.g.
working groups in recent years, and reviews have 19-nortestosterone injected intramuscularly as its
been published (Schnzer and Donike 1993; Schnzer undecanoate ester (Deca-Duraboline), are detectable
1996; Thevis and Schnzer 2010). Basically, the in urine for several weeks; whereas most oral prepar-
metabolism of AAS follows the metabolic pathways ations are completely eliminated within a few days
of the principal androgen testosterone. This includes after intake. Once they became aware of these scien-
reduction of the double bond at C4-C5 to form 5a- tific data, athletes switched their doping activities to
and 5b-isomers, the reduction of the 3-keto group to AAS with short elimination times and to steroids
a 3a-hydroxy function and, in the case of 17b- which were believed to be undetectable.
hydroxy steroids with a secondary hydroxy group,
the oxidation yielding a 17-keto function. Addition- 24.6.2.3 Sample preparation
ally, many AAS are metabolized by cytochrome P450 For sample preparation of anabolic steroids it has to
hydroxylation reactions, and steroids with hydroxy be considered that most of the AAS and their metab-
groups mainly at C-6b, C-16a and C-16b are pro- olites are excreted in conjugated form. Following
duced. In the metabolism of stanozolol, a synthetic sample preparation, unconjugated steroids can be
steroid with a condensed pyrazol ring on the steroid separated by extracting an aliquot of urine (e.g. 2
A-ring (Fig. 24.1), further hydroxylation occurs at ml) with a polar, organic, non-water-miscible solvent.
C-4b and C-30 of the heterocyclic ring. In general, in Based on their polar and acidic character, conjugated
the course of phase I metabolism steroids are enzy- steroids are not extractable and remain in the aqueous
matically transformed to more polar but pharmaco- layer. These conjugates (mainly glucuronides) can be
logically inactive compounds. Phase I reactions are liberated by enzymatic hydrolysis of the urine speci-
often followed by phase II processes, also known as men. The enzyme used can be added directly to the
phase II conjugation. In the case of AAS and their urine or to an isolate obtained via an absorber resin.
metabolites the reaction creates steroid conjugates Enzymatic hydrolysis is achieved completely using
with sulfate or glucuronic acid (Thevis et al. 2001). enzyme preparations with b-glucuronidase from
These highly polar compounds are then rapidly elim- Escherichia coli or b-glucuronidase/arylsulfatase from
inated in the urine. Helix pomatia. The free steroids are then extracted
In the last 20 years excretion studies performed from the aqueous phase via a simple liquid extraction
with 17a-methyl steroids have demonstrated that the with tert-butyl methyl ether, or in the case of less
metabolism of AAS is highly complex, and the detec- polar steroids, with an alkane (e.g. n-pentane).
tion of more than 20 metabolites after administration The first analysis is a screening procedure by
of one single AAS is not unusual. Similar results which all banned AAS are detected in one single

525
Chapter 24: Detecting illegal use of androgens and SARMs

analytical run. Suspicious samples are confirmed an athlete constitutes the misuse of a banned steroid.
by a second aliquot of the same urine specimen, The criteria for identification of a substance are based
which is isolated using a substance-specific isolation on the analytical method applied.
technique. In GC-MS, GC-MS/MS and LC-MS/MS identifi-
cation of synthetic AAS obtained from a urine speci-
24.6.2.4 Derivatization men, it is mandatory to register a full mass spectrum,
For GC-MS analysis based on the polar groups of or a selected ion monitoring (SIM) profile of the main
AAS (hydroxy and keto groups), high interactions abundant fragment ions, or daughter ions generated
with polar functions of the gas chromatography by tandem MS experiments of a diagnostic precursor
column phase reduce the detectability of AAS at low ion (SRM, selected reaction monitoring). The mass
concentrations. Derivatization of polar functions of spectrometric data (MS spectrum, SIM profile or
AAS can lead to a distinct improvement in peak daughter ions) of the isolated substance should be in
intensity and detection limit of the analytical method. accordance with an authentic synthesized reference
The most frequently used derivatization methods are substance or, in the event that a synthesized reference
acylation (e.g. trifluoroacetylation) and silylation (e.g. metabolite is not available, with a well-characterized
trimethylsilylation). For doping analysis of AAS, sily- metabolite from an excretion study with the corres-
lation is the method of choice, and the introduction of ponding AAS. In addition to the MS data, the gas
a trimethylsilyl group to an AAS is the most common chromatography or liquid chromatography retention
derivatization reaction, converting polar groups such time of the isolated steroid has to be in accordance
as hydroxy and keto functions to less polar trimethyl- with the gas chromatography or liquid chromatog-
silyl ethers with excellent gas chromatography behav- raphy retention time of the reference substance. For
ior. For this kind of derivatization a respectable this purpose, reference metabolites of AAS frequently
reagent MSTFA (N-methyl-N-trimethylsilyltrifluoro- misused but not commercially available were synthe-
acetamide) was developed (Donike 1969). Addition- sized (Schnzer and Donike 1993).
ally, the mass spectrum is generally changed to higher As an example, Fig. 24.4 shows the criteria for a
and more abundant molecular and fragment ions, positive sample for a long-term excreted metabolite of
which also improves the signal-to-noise ratio of the metandienone:
substance to be identified compared to the analytical  registration of a full mass spectrum which can be
and biological background. Therefore, derivatization compared with the reference spectrum; or
for GC-MS and GC-MS/MS detection of substances  in case of low concentrations, a selected ion
isolated from biological fluids unequivocally yields a monitoring (SIM) profile with the main
more accurate analytical result, which is an absolute intense fragment ions of the metandienone
requirement in view of the complex matrix and large metabolite 17,17-dimethyl-18-nor-5b-androst-
number of possible interferences. 1,13-dien-3a-ol.
To increase the efficiency of AAS misuse testing and
24.6.3 Detection of synthetic anabolic to detect AAS for a longer period of time after admin-
androgenic steroids istration, more selective and sensitive MS techniques
In some instances AAS are differentiated into have been used during the last decade. The main
endogenous and exogenous AAS according to their improvements were, first, installation of more sensi-
route of administration. The term synthetic should tive and selective mass spectrometers, and second, by
exemplify the fact that these AAS are not produced in substance-specific sample preparation (Schnzer et al.
the body: they are chemically synthesized and can 1996). The use of high-resolution mass spectrometry
only enter the circulating blood system by exogenous (HRMS) was announced to the public at the 1996
application. Anabolic androgenic steroids which are Olympic Games in Atlanta. This technique was estab-
naturally synthesized in the glands of mammalian lished after 1992 in a few IOC-accredited laboratories.
cells are called endogenous steroids, even though their The advantage of HRMS became apparent before
application can be exogenous. As synthetic AAS and/ Atlanta when, during doping testing by the Inter-
or their metabolites are not present in the human national Weightlifting Federation, more than 40 ath-
organism, their identification in a urine sample of letes were confirmed positive only by HRMS and not

526
Chapter 24: Detecting illegal use of androgens and SARMs

(A) 253
100
CH3

CH3
80

CH3
60
H3C Si O
CH3 H
216
40
201
358
185
105 268
20 73 147 163
91 343

50 100 150 200 250 300 350

(B) (C)
Positive sample Reference standard
E+04 m/z: 216.1878 E+04
m/z: 216.1878 6:32 38 %
38 % 4.391 100 4.716

6:24 50

m/z: 253.1956 100 % E+04 m/z: 253.1956 100 % E+05


7.239 100 1.236
50

m/z: 268.2191 16% E+04 m/z: 268.2191 17 % E+04


1.155 100 2.116
50
6:23

m/z: 343.2457 5% E+03 m/z: 343.2457 6% E+03


3.856 100 7.218
50

m/z: 358.2692 10 % E+03 m/z: 358.2692 10 % E+04


7.433 100 1.253
50

6:25 6:30 6:35 6:40 6:45 6:50 6:25 6:30 6:35 6:40 6:45 6:50
Fig. 24.4 Criteria for a positive confirmation: (1) The registered EI (electron impact ionization) mass spectrum, e.g. mass spectrum of
an isolated metabolite of metandienone: 17,17-dimethyl-18-nor-5b-androsta-1,13-dien-3a-ol TMS, (A) has to be in accordance with the
mass spectrum of an authentic reference substance, or (2) The main abundant fragment ions of the isolated substance show similar
intensities (B) when selected ion monitoring (SIM) registration is applied in comparison to the intensities of the same fragments of the
reference compound (C).

527
Chapter 24: Detecting illegal use of androgens and SARMs

by the conventional MS technique. Following these ratios of DHT/epitestosterone, DHT/etiocholanolone,


results the IOC decided that it was necessary that 5a-androstane-3a,17b-diol/5b-androstane-3a,17b-
accredited laboratories use more sophisticated equip- diol, and androsterone/etiocholanolone were estab-
ment, such as HRMS or tandem mass spectrometry lished (Kicman et al. 1995; Donike et al. 1995). Now-
(MS-MS). adays suspicious samples are also forwarded to IRMS
analysis to prove the exogenous origin of DHT and/or
its main metabolites.
24.6.4 Detection of endogenous anabolic
androgenic steroids 24.6.4.2 Direct detection method: gas
24.6.4.1 Indirect detection methods chromatographycombustionisotope ratio mass
The misuse of testosterone by athletes is also tested by spectrometry (GC-C-IRMS)
GC-MS analysis of urinary extracts. The MS data The T/E ratio results can be supported by gas
alone does not distinguish exogenous (doping) from chromatographycombustionisotope ratio mass
endogenous origin of testosterone. In 1983 Donike spectrometry (GC-C-IRMS). This method was first
et al. developed a method to calculate urinary excreted introduced by Becchi et al. in 1994 and has been
testosterone by a ratio to 17-epitestosterone. Both adopted by other research groups (Aguilera et al.
isomeric steroid hormones are excreted mainly as 1996; Horning et al. 1997; Shackleton et al. 1997;
glucuronides which are enzymatically hydrolyzed Flenker et al. 2008) with distinct modifications.
before GC-MS analysis. The urinary testosterone/epi- A review of GC-C-IRMS in dope analysis has been
testosterone ratio (T/E ratio) represents a relatively presented by Cawley and Flenker (2008).
constant factor within an individual, and alterations The principle of IRMS is the precise measurement
under physical exercise have not been noted. Exogen- of the 13C/12C isotope ratio of organic compounds.
ous application of testosterone results in an increase This method became practical for trace analysis in
in the urinary concentration of testosterone glucuro- doping control when instruments combining gas
nide, whereas epitestosterone glucuronide is not chromatography and isotope ratio mass spectrometry
influenced. Based on measurements of large reference were developed. Isotopes are elements with the same
groups, Donike proposed a T/E ratio of 6 : 1 as a number of protons but different numbers of neu-
marker to handle a urine specimen suspicious for trons. Carbon occurs in three kinds of isotopes:
12
testosterone misuse. In 2006 the WADA reduced the C (6 protons and 6 neutrons) with a frequency of
ratio to 4 : 1. An increased T/E value (T/E > 4) is not approximately 98.9%, 13C (6 protons and 7 neutrons)
immediately considered as a positive sample. at a rate of 1.1% and 14C (6 protons and 8 neutrons),
Following the WADA rule, the athlete has to be a radioactive isotope with a half-life of 5760 years
further investigated and it has to be determined that (used in determination of age), in traces. In the course
the increased value is not caused by physical or patho- of synthesizing organic compounds, 12C atoms react
logical conditions. In practice this requires several test slightly faster than 13C atoms. This effect results
samples of the athlete and evaluation of previous tests in a reduction of the 13C amount compared to 12C.
in order to establish the athletes individual T/E refer- The 13C/12C ratio is calculated in per mille (delta
13
ence values (subject-based reference values). Add- C (0/00)) relative to a reference gas with a standard-
itionally, analysis of the sample by IRMS (isotope ized 13C/12C ratio. The delta-value becomes more
ratio mass spectrometry) of carbon is applied to prove negative when the 13C portion is reduced, which
whether the 13C/12C ratio of the urinary excreted occurs during synthetic pathways. For isotope meas-
testosterone and its main metabolites is consistent urement, urinary excreted steroids have to be
with the endogenous production of testosterone (see isolated to high purity. For gas chromatographic sep-
Section 24.6.4.2). aration, derivatization is applied using acetylation of
Doping with DHT became public knowledge after steroids with the aim of improving gas chromatog-
the Asian Games in 1994, when seven athletes were raphy peak shape, or analysis refers to the underiva-
tested positive for DHT misuse. The criteria for DHT tized steroids.
doping are also based on statistical methods, and Steroids are separated by gas chromatography
population-based reference values with limits for the followed by complete oxidation to carbon dioxide in

528
Chapter 24: Detecting illegal use of androgens and SARMs

a combustion chamber. The carbon dioxide is then preclinical or clinical trials, and new drug entities
introduced to the mass spectrometer where the exact are continuously published, demonstrating the
masses m/z 44 for 12CO2 and m/z 45 for 13CO2 are unconfined interest in novel anabolic agents allowing
independently registered. For this kind of isotope for a separation of anabolic and androgenic effects.
ratio measurement, a minimum of 510 ng of a Arylpropionamide-derived SARMs represent one of
steroid has to be used to obtain precise data. The the most advanced classes of compounds, which are
13 12
C/ C ratio can be estimated with an accuracy of complemented by bicyclic hydantoin-, quinolinone-,
 0.0002% ( 0.2 per mille to the 13C/12C ratio of the tetrahydroquinolinone-(Nagata et al. 2011), carba-
reference gas). zole-(Miller et al. 2010), as well as 4-aza-steroid-
A direct proof of exogenous testosterone applica- derived compounds (Schmidt et al. 2010) (Fig.
tion is possible as the delta-values are decreased 24.5), as recently reviewed by Mohler et al. (2009).
to 28 ppm after administration, in comparison Selective androgen receptor modulator based drugs
to 18 to 24 ppm (depending on the nutrition of might offer new alternative therapeutic strategies
the athlete) before intake. This method is also effect- towards the treatment of disease-associated muscle
ive when ethnic differences influence testosterone wasting, osteoporosis, anemia and different classifi-
metabolism, e.g. in Asians who have low T/E ratios, cations of muscular dystrophies; however, concerns
and when a testosterone application will not necessar- about an illicit use infiltrating the world of sports
ily exceed the T/E ratio cutoff (de la Torre et al. arose and were supported by recent findings of non-
1997). Exogenous testosterone also influences the approved drug candidates on the black market (The-
13 12
C/ C ratio of the metabolites of testosterone. Based vis et al. 2009; 2011). Consequently, since January
on these data it was proved that precursors within the 2008, the entire class of androgen receptor modula-
synthetic pathway of testosterone, such as pregnane- tors has been prohibited in sports according to the
diol, pregnanetriol (metabolites of progesterone and anti-doping regulations of the WADA (2010),
17a-hydroxyprogesterone) and cholesterol are not although no SARM product has yet been officially
influenced by exogenous testosterone, whereas testos- launched. Nevertheless, such therapeutics may be
terone and its metabolites have decreased delta-values particularly attractive to athletes as they presumably
indicating exogenous application. offer the desired anabolic effects without typical
The IRMS method can additionally be applied to undesirable effects commonly associated with ana-
detect and identify doping with other endogenous bolic androgenic steroid (mis)use.
AAS such as dihydrotestosterone and DHEA.

24.7.2 Analytical approaches


24.7 Selective androgen receptor For sports drug testing purposes, numerous detec-
modulators (SARMs) tion methods have been established during recent
years, and several approaches targeting the intact
24.7.1 General aspects compounds as well as in-vitro, in-vivo or chemically
Selective androgen receptor modulators (SARMs) derived metabolites were reported (Thevis et al.
have received considerable attention from the doping 2006; 2007a; 2008a; 2008b; 2008c; 2010; Thevis and
control community since determined medicinal Schnzer 2008). Most of these methods are based on
research outlined their potential beneficial properties liquid chromatography(tandem) mass spectrometry
for the treatment of various diseases (Gao and (LC-MS(/MS)), as the structural diversity of SARMs
Dalton 2007; Kilbourne et al. 2007; Mohler et al. represents a key factor in sample preparation and
2008; 2009; Bhasin and Jasuja 2009). Due to the analysis of this class of emerging drugs. As depicted
major goal of developing ligands that induce anabol- in Fig. 24.5, all representatives of SARMs comprise
ism specifically in muscle and bone tissue, a misuse polar functions that lead to positive (2-quinolinones)
of SARMs (comparable to the illicit use of steroidal or negative (arylpropionamides, bicyclic hydantoins)
and other non-steroidal agents) in elite and amateur charges during the analytical process. Moreover, the
sport cannot be excluded. A great variety of drug metabolic fate of most compounds is yet to be fully
candidates based on considerably different pharma- elucidated. Solid-phase extraction (SPE) and liquid-
cophores has been prepared and investigated in liquid extraction (LLE) were chosen for first studies

529
Chapter 24: Detecting illegal use of androgens and SARMs

(A) Fig. 24.5 Chemical structures of


(E)
selected representatives of SARMs with
NC arylpropionamide (S-22, A, and Andarine,
O O2N
O CN B), bicyclic hydantoin (BMS-564929, C),
CF3 N quinoline (LGD-2226, D),
HH C OH N OH tetrahydroquinoline (E), carbazole
3 H
H3C CH3 (RAD35101, F), and 4-aza-steroid
(B) (MK-0773, G) -derived nuclei.

O 2N (F)
O
H H
O N Cl CF3
N
CF3 N O OH
HH C OH
3 H3C
Cl
(C)
HO O

N N O H N
N (G) N N
CH3 N
O H3C Cl H
CH3
(D) F
CF3
CF3
N CF3 O N
CH3

O N
H

on the detection of SARMs in doping control analy-


sis, followed by LC-ESI-MS/MS (liquid chromatog- 24.8 Outlook
raphy with electrospray ionizationtandem mass Improvement in the fight against doping was
spectrometry) analysis, which enabled the detection achieved by: national and international founding of
of 1 ng/ml of arylpropionamide-derived SARMs national anti-doping agencies and the World Anti-
from urine specimens. The simultaneous measure- Doping Agency independent of sport organizations;
ment of precursor ion scans on most common prod- the development and improvement of more efficient
uct ions derived from the conserved nucleus, or high detection methods supported by national and inter-
resolution/high accuracy MS targeting these core frag- national research grants; the legalization of anti-
ment ions following all ion fragmentation (e.g. by doping actions by national laws to allow police
in-source collision-induced dissociation), comple- investigations.
mented the assay to allow for a comprehensive For analysis of AAS, the general strategy is to
screening for related drugs as well as their metabolites improve sensitivity to detect exogenous AAS, by
(Thevis et al. 2006; Thomas et al. 2010). The bicyclic GC-MS(MS) or LC-MS(MS), possibly several weeks
hydantoin BMS-564929 (Fig. 24.5C) was also after the last application. Currently this appears to be
extracted from spiked urine samples using SPE, but the best approach, as the efficiency of testing pro-
due to a poorly acidic nature and limited proton grams for athletes in out-of-competition periods in
affinity, methanol adduct formation under negative countries with less democratic structures is
ESI conditions was employed to allow detection limits questionable.
of 520 ng/ml (Thevis et al. 2007b). In contrast, 2- For the misuse of endogenous AAS, a program is
quinolinone-derived SARMs (Fig. 24.5D) were isol- planned by WADA referred to as The Athletes Bio-
ated from urine specimens by means of common logical Passport. This program is a follow-up of
LLE, and LC-MS/MS using positive ESI enabled the continuous testing of athletes to establish their indi-
detection of 0.2 ng/ml (Thevis et al. 2007a). vidual steroid profile data, in general, for testosterone

530
Chapter 24: Detecting illegal use of androgens and SARMs

and its main metabolites. This program is not entirely 24.9 Key messages
new as the T/E ratio testing has been well established
 Misuse of androgens in competitive sports has
for over 20 years. The program will cover all athletes, been banned since 1974 and is tested by
not only athletes with elevated T/E ratio, i.e. greater WADA-accredited laboratories.
than four.  Androgens are used by athletes during training
The implementation of new technologies espe- to improve muscle strength.
cially in MS has yielded improvements in selectivity  Non-therapeutic hormones such as
and sensitivity for the detection of banned substances. prohormones of testosterone and
Beside the use of urine as biological matrix, current nortestosterone and designer steroids have
tests are including more and more blood samples. been marketed as nutritional supplements since
Methods for hair analysis of AAS have been 1999.
 Positive doping cases have been proved to
developed and may be used in the future, too. Other
originate from the use of nutritional supplements
matrices such as saliva or even breath may be of contaminated with prohormones of
interest as well. Preventive doping research may focus nortestosterone.
on these matrices, keeping in mind the strong  Testosterone, nortestosterone, stanozolol and
improvements in MS. metandienone represent the most frequently
The fight against doping, which has been in the misused AAS in controlled sports.
hands of sport organizations for more than 40 years,  Synthetic androgens are extensively metabolized,
was improved when several countries implemented and doping tests are focused on urinary excreted
independent anti-doping agencies and set in force metabolites.
anti-doping laws which allow police investigations in  Doping with endogenous steroids is controlled
the case of an anti-doping rule violation in sport, or in by indirect methods; e.g. testosterone misuse is
tested by a ratio of testosterone to
case of strong suspicions of practicing with doping
epitestosterone (4 : 1). Samples with elevated
substances or doping methods. In general such regu- ratios are followed up by additional studies to
lations prohibit the distribution of doping substances exclude physiological and pathological
and methods to athletes. The aim is not to punish the influences.
athletes but those persons responsible for providing  Direct methods, such as gas
the banned substances, e.g. trainers, physicians, other chromatographycombustioncarbon isotope
individuals of the healthcare system and dealers. ratio mass spectrometry have become available
However, the key elements in the international to identify doping with endogenous steroids
anti-doping fight remain the doping control of ath- unambiguously.
letes, including chemical analysis of samples. Support  Selective androgen receptor modulators are
by national anti-doping rules is appreciated, and designed to tissue-selectively stimulate or inhibit
the androgen receptor.
police investigations can help to provide additional
 Due to their non-steroidal structure, SARMs are
data to support actions against doping structures not subject to metabolic degradation by
independent of anti-doping sport regulations. These typical steroid-metabolizing enzymes such as
should be considered as synergistic measures to 5a-reductase.
improve the anti-doping fight in sport.

24.10 References P (1994) Gas chromatography/


combustion/isotope-ratio mass
nortestosterone esters in normal
men. J Steroid Biochem 22: 623629
Aguilera R, Becchi M, Casabianca H, spectrometry analysis of urinary
Hatton CK, Catlin DH, Starcevic B, Bhasin S, Jasuja R (2009) Selective
steroids to detect misuse of androgen receptor modulators as
Pope HG Jr (1996) Improved testosterone in sport. Rapid
method of detection of function promoting therapies. Curr
Commun Mass Spectrom 8: 304308 Opin Clin Nutr Metab Care 12:
testosterone abuse by gas
chromatography/combustion/ Beckett AH, Cowan DA (1979) Misuse 232240
isotope ratio mass spectrometry of drugs in Sport. Brit J Sports Med
Bhasin S, Storer TW, Berman N,
analysis of urinary steroids. J Mass 2: 185194
Callegari C, Clevenger B, Phillips J,
Spectrom 31: 169176 Belkien L, Schurmeyer T, Hano R, Bunell TJ, Tricker R, Shirazi A,
Becchi M, Aguilera R, Farizon Y, Gunnarsson PO, Nieschlag E (1985) Casaburi R (1996) The effects of
Flament MM, Casabianca H, James Pharmacokinetics of 19- supraphysiologic doses of

531
Chapter 24: Detecting illegal use of androgens and SARMs

testosterone on muscle size and Leistung und Gesundheit. Deutscher Horning S, Geyer H, Machnik M,
strength in normal men. J Med 335: rzte Verlag, Cologne, pp 293298 Schnzer W, Hilkert A, Oeelmann
17 Donike M, Zimmermann J, Brwald J (1997) Detection of exogenous
Boos C, Wulff P, Kujath P, Bruch HP KR, Schnzer W, Christ V, testosterone by 13C/12C analysis.
(1998) Medikamentenmissbrauch Klostermann K, Opfermann G In: Schnzer W, Geyer H,
im Freizeitsport. Deutsches (1984) Routine Bestimmung von Gotzmann A, Mareck-Engelke U
rzteblatt 95(16): A953A957 Anabolika im Harn. Deutsch (eds) Proceedings of the 14th Cologne
Z Sportmed 1: 1423 Workshop on Dope Analysis 1996.
Brooks RV, Firth R, Summer NA Sport und Buch Strau, Cologne,
(1975) Detection of anabolic Donike M, Ueki M, Kuroda Y, pp 275283
steroids by radio immunoassay. Br Geyer H, Nolteernsting E, Rauth S,
J Sports Med 9: 8992 Schnzer W, Schindler U, Vlker E, Kamber M (1995) Mitteilung in
Fujisaki M (1995) Detection of Doping Information und
Canadian Centre for Drug-Free Sport Prvention. Maggelingen 7: 47
(1993) News Release Over 80,000 dihydrotestosterone (DHT) doping:
young Canadians using anabolic alteration in the steroid profile and Kazlauskas R (2010) Designer steroids.
steroids. CCDFS, Montreal reference ranges for DHT and its In: Thieme D, Hemmersbach P
5a-metabolites. J Sports Med Phys (eds) Doping in Sports. Handbook of
Catlin DH, Ahrens BD, Kucherova Y Fitness 35: 235250 Experimental Pharmacology, Vol.
(2002) Detection of norbolethone,
Duchaine D (1989) Underground 195, Springer, Heidelberg, pp
an anabolic steroid never marketed,
Steroid Handbook II. Technical 155185
in athletes urine. Rapid Commun
Mass Spectrom 16: 12731275 Books, Venice, FL Kicman AT, Coutts SB, Walker CJ,
Catlin DH, Sekera MH, Ahrens BD, Flenker U, Gntner U, Schnzer W Cowan DA (1995) Proposed
Stracevic B, Chang YC, Hatton CK (2008) Delta13C-values of confirmatory procedure for
(2004) Tetrahydrogestrinone: endogenous urinary steroids. detecting 5a-dihydrotestosterone
discovery, synthesis, and detection Steroids 73: 408416 doping in male athletes. Clin Chem
in urine. Rapid Commun Mass Franke WW, Berendonk B (1997) 41: 16171627
Spectrom 18: 12451249 Hormonal doping and Kicman AT, Bassindale T, Cowan DA,
Cawley AT, Flenker U (2008) The androgenization of athletes: a secret Dale S, Hutt AJ, Leeds AR (2003)
application of carbon isotope ratio program of the German Democratic Effect of androstenedione ingestion
mass spectrometry to doping Republic government. Clin Chem on plasma testosterone in young
control. J Mass Spectrom 43: 43: 12621279 women; a dietary supplement with
854864 Gao W, Dalton JT (2007) Expanding potential health risks. Clin Chem 49:
the therapeutic use of androgens via 167169
Clasing D (1992) Doping verbotene
Arzneistoffe im Sport. Gustav selective androgen receptor Kilbourne EJ, Moore WJ, Freedman
Fischer Verlag, Stuttgart modulators (SARMs). Drug Discov LP, Nagpal S (2007) Selective
Today 12: 241248 androgen receptor modulators for
de la Torre X, Segura J, Yang Z, Li Y, frailty and osteoporosis. Curr Opin
Wu M (1997) Testosterone Geyer H, Parr MK, Mareck U, Reinhart
U, Schrader Y, Schnzer W (2004) Investig Drugs 8: 821829
detection in different ethnic groups.
In: Schnzer W, Geyer H, Analysis of non-hormonal Kochakian CD (1976) Anabolic-
Gotzmann A, Mareck-Engelke U nutritional supplements for Androgenic Steroids. Springer-
(eds) Proceedings of the 14th Cologne anabolic-androgenic steroids Verlag, Berlin
Workshop on Dope Analysis 1996. results of an international study. Int Kochakian CD (1990) A steroid review:
Sport und Buch Strau, Cologne, J Sports Med 25: 124129 metabolite of testosterone;
pp 7189 Grundig P, Bachmann M (1995) World significance in the vital economy.
Donike M (1969) N-Methyl-N- Anabolic Review 1996. Sport Verlag Steroids 55: 9297
trimethylsilyl-trifluoracetamid, ein Ingenohl, Heilbronn Leder BZ, Longcope C, Catlin DC,
neues Silylierungsmittel aus der Handelsman DJ, Gupta L (1997) Ahrens B, Schoenfeld DA,
Reihe der silylierten Amide. Prevalence and risk factors for Finkelstein JS (2000) Oral
J Chromatogr 42: 103104 anabolic-androgenic steroid abuse androstenedione administration
Donike M, Brwald KR, Klostermann in Australian high school students. and serum testosterone
K, Schnzer W, Zimmermann J Int J Androl 20: 159164 concentrations in young men.
(1983) Nachweis von exogenem Haupt HA, Rovere GD (1984) Anabolic JAMA 283: 779782
Testosteron. In: Heck H, Hollmann W, steroids: a review of the literature. Mass R, Ayotte C, Dugal R (1989)
Liesen W, Rost R (eds) Sport: Am J Sports Med 12: 469484 Integrated methodological approach

532
Chapter 24: Detecting illegal use of androgens and SARMs

to the gas chromatographic mass stanozolol abuse in athletes by gas Striegel H, Simon P, Frisch S, Roecker
spectrometric analysis of anabolic chromatography/high resolution K, Dietz K, Dickhuth HH, Ulrich R
steroid metabolites in urine. mass spectrometry (GC/HRMS). (2006) Anabolic ergogenic
J Chromatogr 489: 2350 J Chormatogr B 687: 93108 substance users in fitness-sports: a
Miller CP, Bhaket P, Muthukaman N, distinct group supported by the
Schnzer W, Geyer H, Fuhller G,
Lyttle CR, Shomali M, Gallacher K, health care system. Drug Alcohol
Halatcheva N, Kohler M, Parr MK,
Slocum C, Hattersley G (2010) Depend 81: 1119
Guddat S, Thomas A, Thevis M
Synthesis of potent, substituted (2006) Mass spectrometric Taylor WN (1982) Steroids and the
carbazoles as selective androgen identification and characterization Athlete. McFarland & Company,
receptor modulators (SARMs). of a new long-term metabolite of London
Bioorg Med Chem Lett 20: metandienone. Rapid Commun Thevis M, Schnzer W (2008) Mass
75167520 Mass Spectrom 10: 22522258 spectrometry of selective androgen
Mohler ML, Bohl CE, Narayanan R, Schmidt A, Kimmel DB, Bai C, receptor modulators. J Mass
He Y, Hwang DJ, Dalton JT, Miller DD Scafonas A, Rutledge S, Vogel RL, Spectrom 43: 865876
(2008) Nonsteroidal tissue-selective McElwee-Witmer S, Chen F, Thevis M, Schnzer W (2010) Synthetic
androgen receptor modulators. In: Nantermet PV, Kasparcova V, Leu anabolic agents: steroids and
Ottow E, Weinmann H (eds) CT, Zhang HZ, Duggan ME, nonsteroidal selective androgen
Nuclear Receptors as Drug Targets. Gentile MA, Hodor P, Pennypacker receptor modulators. In: Thieme D,
Wiley-VCH, Weinheim, B, Masarachia P, Opas EE, Adamski Hemmersbach P (eds) Doping in
pp 249304 SA, Cusick TE, Wang J, Mitchell HJ, Sports. Handbook of Experimental
Mohler ML, Bohl CE, Jones A, Coss Kim Y, Prueksaritanont T, Perkins Pharmacology, Vol. 195. Springer,
CC, Narayanan R, He Y, Hwang DJ, JJ, Meissner RS, Hartman GD, Heidelberg, pp 99126
Dalton JT, Miller DD (2009) Freedman LP, Harada S, Ray WJ
Thevis M, Opfermann G, Schmickler
Nonsteroidal selective androgen (2010) Discovery of the selective
H, Schnzer W (2001) Mass
receptor modulators (SARMs): androgen receptor modulator MK-
spectrometry of steroid glucuronide
dissociating the anabolic and 0773 using a rational development
conjugates. I. Electron impact
androgenic activities of the strategy based on differential
fragmentations of 5a-/5b-
androgen receptor for therapeutic transcriptional requirements for
androstan-3a-ol-17-one
benefit. J Med Chem 52: 35973617 androgenic anabolism versus
glucuronides, 5a-estran-3a-ol-17-
Nagata N, Miyakawa M, Amano S, reproductive physiology. J Biol
one glucuronide and deuterium-
Furuya K, Yamamoto N, Inoguchi Chem 285: 1705417064
labelled analogues. J Mass Spectrom
K (2011) Design and synthesis of Schrader Y, Thevis M, Schnzer W 36: 159168
tricyclic tetrahydroquinolines as a (2006) Quantitative determination Thevis M, Kamber M, Schnzer W
new series of nonsteroidal selective of metabolic products of 19-
(2006) Screening for metabolically
androgen receptor modulators norandrostenediol in human stable aryl-propionamide-derived
(SARMs). Bioorg Med Chem Lett 21: plasma using gas chromatography/
selective androgen receptor
17441747 mass spectrometry. Drug Metab modulators for doping control
Parr MK, Schnzer W (2010) Detection Dispos 34: 13281335
purposes. Rapid Commun Mass
of the misuse of steroids in doping Sekera MH, Ahrens BD, Chang YC, Spectrom 20: 870876
control. J Steroid Biochem Mol Biol Starcevic B, Georgakopoulos C,
121: 528537 Thevis M, Kohler M, Maurer J,
Catlin DH (2005) Another designer Schlrer N, Kamber M, Schnzer W
Schnzer W (1996) Review steroid: discovery, synthesis, and (2007a) Screening for 2-
metabolism of anabolic androgenic detection of madol in urine. Rapid quinolinone-derived selective
steroids. Clin Chem 42: 10011020 Commun Mass Spectrom 19: androgen receptor agonists in
Schnzer W, Donike M (1993) 781784 doping control analysis. Rapid
Metabolism of anabolic steroids in Shackleton CH, Phillips A, Chang T, Li Commun Mass Spectrom 21:
man: synthesis and use of reference Y (1997) Confirming testosterone 34773486
substances for identification of administration by isotope ratio Thevis M, Kohler M, Schnzer W
anabolic steroid metabolites. Anal mass spectrometric analysis of (2007b) Mass spectrometry of new
Chim Acta 275: 2348 urinary androstanediols. Steroids growth promoting drugs:
Schnzer W, Delahaut P, Geyer H, 62: 379387 hydantoin-derived selective
Machnik M, Horning S (1996) Sinner D (2009) Anabole Steroide Das androgen receptor modulators and
Longterm detection and Schwarze Buch 2010. BMS Verlag, growth hormone secretagogues. In:
identification of metandienone and Gronau, Germany Schnzer W, Geyer H, Gotzmann A,

533
Chapter 24: Detecting illegal use of androgens and SARMs

Mareck U (eds) Recent Advances in androgen receptor modulator Prohibited-list/


Doping Analysis. Sport und Buch (SARM) S-4 (Andarine) in a black- WADA_Prohibited_List_2010_EN.
Strau, Cologne, pp 263272 market product. Drug Test Anal 1: pdf (accessed Jan 14, 2010)
Thevis M, Kohler M, Schlrer N, 387392 Wade N (1972) Anabolic steroids:
Fusshller G, Schnzer W (2008a) Thevis M, Thomas A, Fusshller G, doctors denounce them, but athletes
Screening for two selective Beuck S, Geyer H, Schnzer W arent listening. Science 176:
androgen receptor modulators (2010) Mass spectrometric 13991403
using gas chromatography-mass characterization of urinary Ward RJ, Lawson AM, Shackleton CHL
spectrometry in doping control metabolites of the selective (1975) Screening by gas
analysis. Eur J Mass Spectrom androgen receptor modulator chromatographymass
(Chichester, Eng) 14: 153161 andarine (S-4) for routine doping spectrometry for metabolites of five
Thevis M, Kohler M, Thomas A, control purposes. Rapid Commun commonly used anabolic steroid
Schlrer N, Schnzer W (2008b) Mass Spectrom 24: 22452254 drugs. Br J Sports Med 9: 9397
Doping control analysis of tricyclic Thevis M, Geyer H, Thomas A, Wilson JD (1988) Androgen abuse by
tetrahydroquinoline-derived Schnzer W (2011) Trafficking of athletes. Endocr Rev 9: 191199
selective androgen receptor drug candidates relevant for sports
modulators using liquid drug testing: detection of non- Wilson JD (1996) Androgens. In:
chromatography/electrospray approved therapeutics categorized Hardmann JG, Limbird IE,
ionization tandem mass as anabolic and gene doping agents Molinoff PB, Ruddon RW,
spectrometry. Rapid Commun Mass in products distributed via the Goodman Gilman A (eds)
Spectrom 22: 24712478 Internet. Drug Test Anal 3: 331336 Goodman and Gilmans The
Pharmacological Basis of
Thevis M, Lohmann W, Schrader Y, Thomas A, Guddat S, Kohler M, Krug Therapeutics, 9th edn. McGraw Hill,
Kohler M, Bornatsch W, Karst U, O, Schnzer W, Petrou M, Thevis M New York, pp 14411457
Schnzer W (2008c) Use of an (2010) Comprehensive plasma-
electrochemically synthesised screening for known and unknown Yesalis CE, Kennedy NJ, Kopstein AN,
metabolite of a selective androgen substances in doping controls. Bahrke MS (1993) Anabolic-
receptor modulator for mass Rapid Commun Mass Spectrom 24: androgenic steroid use in the United
spectrometry-based sports drug 11241132 States. JAMA 270: 12171221
testing. Eur J Mass Spectrom WADA (2010) The 2010 Prohibited Zimmermann J (1986) Untersuchungen
(Chichester, Eng) 14: 163170 List. World Anti-Doping Agency, zum Nachweis von exogenen Gaben
Thevis M, Geyer H, Kamber M, Montreal. Available at: www.wada- von Testosterone. Thesis, German
Schnzer W (2009) Detection of the ama.org/Documents/World_Anti- Sports University, Cologne.
arylpropionamide-derived selective Doping_Program/WADP- Hartung-Gorre Verlag, Konstanz

534
Chapter
Sequelae of doping with anabolic steroids

25 Elena Vorona and Eberhard Nieschlag

25.1 Introduction: the dimension of the 25.2.9 Causes of sudden death under anabolic
problem 535 androgenic steroids 539
25.2 Side-effects of anabolic androgenic steroids in 25.3 Specific side-effects in men 540
both sexes 536 25.4 Effects of anabolic androgenic steroids in
25.2.1 Skin 536 women 540
25.2.2 Blood 537 25.4.1 Hirsutism 541
25.2.3 Heart and vessels 537 25.4.2 Deepening of the voice 541
25.2.4 Liver 538 25.4.3 The question of breast cancer 541
25.2.5 Bones and muscles 538 25.5 Conclusion 542
25.2.6 Kidneys 539 25.6 Key messages 542
25.2.7 Tumors 539 25.7 References 543
25.2.8 Behavior 539

25.1 Introduction: the dimension of In sharp contrast to the great number of AAS users
stands the fact that the possible side-effects of AAS
the problem have not been evaluated in randomized controlled
Since ancient times substances have been consumed and trials (RCTs), which are the gold standard in modern
methods applied to enhance physical performance in evidence-based medicine. There are various reasons
athletic activities. Since the 1950s anabolic androgenic for this. None of the AAS (nor any other substance)
steroids (ASS) have played a dominant role in doping, has been approved by regulatory agencies for the
the modern designation for such generally unsanctioned purpose of doping and therefore subjected to the
manipulations. Although many other hormones and usual regulatory scrutinization which requires proper
drugs are also used for doping, e.g. GH, hCG, thyroid toxicology and RCTs. Anabolic androgenic steroid
hormones, erythropoietin, SARMs, amphetamines, b- doses used for doping are 5 to 20 times greater than
receptor agonists, diuretics, cocaine etc., and gene doping clinically applied doses i.e. doses which would only
is on the horizon, AAS still dominate the field (see be used during drug development in toxicological
Chapter 24). AAS are not only used in high-performance animal studies but not in clinical trials. As the use of
competitive athletics, but also in amateur and popular AAS is illicit, it would be extremely difficult to enroll
sports, fitness training and bodybuilding, so that the current users in RCTs as they would not admit
number of users is astonishingly high, indicating that AAS use and would reject revealing their identity. In
AAS indeed produce the desired effects, perceived or addition, most AAS users also simultaneously admin-
real, in the consumer (Hartgens and Kuipers 2004). ister other drugs, often of unknown purity and
As there are no effects of medications without at undefined doses. This makes it very difficult to
side-effects, it is not surprising that a number of establish any causal relationship between a specific
unwanted sequelae are attributed to the use of AAS. substance and side-effects. This is illustrated by a

Testosterone: Action, Deficiency, Substitution, ed. Eberhard Nieschlag and Hermann M. Behre, Assoc. ed. Susan Nieschlag.
Published by Cambridge University Press. Cambridge University Press 2012.

535
Chapter 25: Sequelae of doping with anabolic steroids

27-year old Olympic athlete who died in 1987 from and the dose applied. For example, it has been shown
toxic multiple organ failure: 102 drugs were identified that changes in testosterone levels in men were asso-
in her body; among others, stanozolol, aspirin, diclo- ciated with dose-dependent and region-specific
fenac, metamizol, codeine and heparin (Fischer- changes in adipose tissue and lean body mass in
Solms 2009). Retrospective systematic analyses are the extremities and trunk (Woodhouse et al. 2004).
also lacking, as, again, former consumers do not come Similarly, side-effects of clinical doses of testoster-
forward, especially if they were champions and medal one are modulated by the polymorphism of the AR
winners, except in a few rare cases. Often they also (Zitzmann and Nieschlag 2007). Supraphysiological
just forget what they were taking and at what doses; or serum testosterone concentrations not only reduced
they never knew. subcutaneous fat, but led to a significant loss of adi-
In the former German Democratic Republic pose tissue from the smaller, deeper intermuscular fat
(GDR), a systematic nationwide doping program stores (Bhasin et al. 1996).
existed for high-performance athletics. The major Another critical question is whether the influences
anabolic androgenic steroid used in that program was of AAS are the same in male and female athletes.
chlordehydromethyltestosterone (Oral Turinabol). Except for the typical sex-dependent effects (e.g. dys-
This had primarily an anabolic effect, which leads menorrhea, clitoral hypertrophy and breast atrophy
to a distinct gain of muscle mass during intensive in women, and testicular atrophy, azoospermia and
training. Its only slight androgenic effect explained its gynecomastia in men), it is generally assumed that
widespread use by female athletes. The active ingredi- AAS act similarly in men and women, but this
ent is metabolized in the body quite rapidly and was remains unclear.
not detectable in blood in doping tests five to seven The following summary of side-effects is based on
days after withdrawal of AAS. Production of endogen- extrapolation from effects observed in patients treated
ous testosterone in males during its use was not sig- with AAS and on descriptions of individual cases or
nificantly influenced, and was again fully intact a short groups of cases, mainly retrospective and hardly ever
time after discontinuation of the drug. The normal controlled.
dose in male athletes was 2040 mg/day; females took
520 mg/day. Some athletes took considerably higher
amounts of the agents (up to 100 mg daily; Sinner and 25.2 Side-effects of anabolic
Bachmann 2004). androgenic steroids in both sexes
The full dimension of this program became
evident only after the collapse of the regime in 1989 25.2.1 Skin
(Berendonk 1991; Franke and Berendonk 1997; Anabolic androgenic steroids act through the AR, pre-
Steinigen 2003). It has been estimated that altogether senting in epidermal and follicular keratinocytes, sebo-
about 10 000 athletes were involved in this program. cytes, sweat gland cells, dermal papilla cells, dermal
In 2002 the German Federal Parliament passed a law fibroblasts, endothelial cells and genital melanocytes.
(Dopingopferhilfegesetz; DOHG) to financially com- The use of AAS can very rapidly lead to cutaneous
pensate former GDR athletes for any demonstrable changes in previously unaffected athletes through
physical damage caused by doping. This situation in affection of e.g. sebaceous gland growth and differen-
Germany is unique insofar as a state-supported pro- tiation, hair growth, epidermal barrier homeostasis
gram organized the doping. Affected athletes now and wound healing (Zouboulis et al. 2007). The AR
demand compensation from the Federal Republic of polymorphism appears to play a role in the severity of
Germany as the legal successor of the GDR. This the symptoms (Zouboulis et al. 2007).
places the athlete in an awkward position, as he/she The most frequent skin manifestations are acne
needs to demonstrate causal relationships between vulgaris, oily skin, seborrhea, striae, hirsutism and
doping and possible disease; particularly because of male pattern alopecia (Walker and Adams 2009).
the absence of any systematic studies concerning the Over 50% of athletes taking part in a questionnaire
long-term effects of doping, in particular AAS. aiming to identify unsupervised AAS regimens and
Furthermore the effects and side-effects of AAS side-effects of AAS reported acne (Evans 1997). After
are dependent on the genetic makeup of the user as elimination of the causal agent these changes are
well as on body weight, duration of administration mostly reversible. To speed up recovery antiandrogen

536
Chapter 25: Sequelae of doping with anabolic steroids

therapy with cyproterone acetate or spironolactone polymorphism that modifies the erythropoiesis-
might be tried (Zouboulis et al. 2007). However, stimulating effect of testosterone in substituted
severe forms of anabolic androgenic steroid-induced patients is of influence in athletes is not known
acne conglobata will leave extensive scarring on the (Zitzmann and Nieschlag 2007).
affected skin areas (Gerber et al. 2008).
After acne, striae distensae as a result of rapid
muscular hypertrophy, supported by AAS intake, 25.2.3 Heart and vessels
is the most prevalent skin side-effect in athletes, espe- It has been demonstrated that long-term AAS users
cially in bodybuilders. Over 40% of athletes com- had an altered electrophysiological capacity of the
plained about stretch marks of the skin (Parkinson myocardium, with a significantly higher incidence
and Evans 2006), with typical localization in the mus- of abnormal electrocardiograms post-exercise (e.g.
culus pectoralis or upper arm region. After discon- extension of QRS complex >114 ms, arrhythmias,
tinuation of drug misuse striae can persist as white including atrial fibrillation, ventricular fibrillation,
streaks (Wollina et al. 2007). ventricular tachycardia, supraventricular and ven-
tricular ectopic beats) compared with controls (Achar
et al. 2010; Sculthorpe et al. 2010).
25.2.2 Blood Anabolic androgenic steroids can often cause con-
Administration of testosterone and anabolic steroids centric left ventricular myocardial hypertrophy, the
to healthy men causes temporary activation of the extent of which seems to be dose related (Dickerman
coagulation system as well as of fibrinolysis. Both et al. 1998; Karila et al. 2003). It has been shown that
changes were reversed after discontinuation of the AAS exert a long-standing hypertrophic effect on the
drug (Kahn et al. 2006). For example, testosterone myocardium. Here there are no significant differences
can increase the activity of thromboxanA2 receptors between current and previous AAS users (De Piccoli
and thrombocyte aggregation, and thereby also the et al. 1991; Di Bello et al. 1999; Vogt et al. 2002).
risk of thrombosis. Simultaneously the activity of the Anabolic androgenic steroids seem not to influence
fibrinolytic system rises, in particular of antithrombin systolic heart function. However, as anabolics affect
III and of protein S (Ferenchick et al. 1995; Shapiro the diastolic function of the left ventricle, this serves
et al. 1999). Levels of plasmina2-antiplasmin com- as a criterion for differentiation between physiologic,
plex (PAP, terminal marker of fibrinolysis), of factor training-induced hypertrophy and a pathological
XIIc and of antithrombin sank significantly in males myocardium (Caso et al. 2006; Kindermann 2006).
who received testosterone undecanoate as depot injec- The athletes heart is characterized by moderate
tions (Zitzmann et al. 2002). Hormone replacement proportional myocardial hypertrophy without func-
therapy containing androgens decreased the plas- tional limitations. Training without AAS does not
minogen-activator-inhibitor-1 (PAI-1) in premeno- lead to thickened heart chamber walls. The patho-
pausal women, leading to improved fibrinolytic logical left ventricular myocardial hypertrophy,
activity (Winkler 1996). developing under AAS intake, is often associated with
Changes of the hemostatic system during testos- restricted diastolic function of the affected heart
terone therapy have also been investigated in female chamber, probably caused by increasing myocardial
transsexuals (female-to-male) who received 250 mg fibrosis. The second diagnostic criterion is the thick-
testosterone enanthate injections every two weeks ness of the left ventricular myocardium seen on echo-
over an extended period of time (Toorians et al. cardiography. A heart chamber wall thickness of
2003). This therapy had a slight antithrombotic effect. more than 13 mm is suspicious of pathological myo-
Anabolic androgenic steroids cause a significant cardial hypertrophy or AAS misuse (Dickerman et al.
increase of erythrocytes and hemoglobin concentra- 1998; Kindermann 2006). A left ventricular hyper-
tion (Aln 1985; Clasing and Mller 2008; Sjqvist trophy can be detected on echocardiography several
et al. 2008; Nieschlag et al. 2010), which constitutes years after AAS withdrawal (Achar et al. 2010).
part of the intended effects as it increases oxygen Although myocardial hypertrophy seems to be revers-
transport. However, increases in the hematocrit ible, impaired diastolic function of the left ventricle
above 52% may lead to thrombosis and apoplexy, and the decreased inotropic capacity of the myocar-
especially in older subjects. To what extent the AR dium are irreversible (Turillazzi et al. 2011).

537
Chapter 25: Sequelae of doping with anabolic steroids

It has been reported that uncontrolled AAS does the lipid status return to normal. After long-
misuse can cause a significantly higher incidence of standing use atherosclerosis and consequential coron-
cardiac death in apparent healthy young athletes. This ary heart disease, cerebral vessel disease or peripheral
concerns mainly powerlifters and bodybuilders taking arterial obstructive disease could hypothetically
very high AAS doses, often as a mixture with other develop, but this has not been proven.
drugs. In his dissertation, Kistler (2006) described It should be remembered here that during the last
effects of AAS misuse on the human organism, based decade a number of studies have appeared which
on autopsy data of 10 young bodybuilders (mean age reverse the role of testosterone from a risk factor for
33.7 years) who took unsupervised mixtures of drugs cardiovascular diseases to a cardioprotective agent
to enhance performance. In four cases the cause of (see Chapter 10). However, this applies to testoster-
death was acute cardiac dysfunction. In all 10 cases one doses in the physiological range and can certainly
the mean heart weight of 517 g was significantly be different if excessive AAS doses are applied.
higher than the mean physiological heart weight.
Furthermore, in all cases chronic ischemic changes 25.2.4 Liver
of the myocardium were found histologically. It is
Changes of the liver structure have been described,
also notable that in almost all cases arteriosclerosis
mainly in cases of chronic misuse of the 17a-
of the coronary vessels and atheromatosis of the
alkylated AAS, e.g. methyltestosterone, metandienone,
arteria carotis and aorta were found, despite the rela-
oxandrolone, stanozolol (Turillazzi et al. 2011). Because
tively young age of the athletes.
of their liver toxicity, 17a-alkylated androgenic steroids
Some cases of dilatative cardiomyopathy have
are considered obsolete for clinical use (at least in
been described in healthy young bodybuilders during
Europe) (Nieschlag 1981), but continue to be available
intake of AAS. In all cases it was uncontrolled high-
illegally for doping purposes. As a direct toxic effect on
dose AAS misuse, particularly in combination with
hepatocytes with ultrastructural cell damage, oxidative
other drugs (Schollert and Bendixen 1993; Clark
stress leading to increased ROS (reactive oxygen
and Schofield 2005). In individuals with a genetic
species) production could play a role in the hepatotoxi-
disposition for dilatative cardiomyopathy using AAS
city of AAS.
it becomes specifically difficult to disentangle causal
Changes often observed are intrahepatic cholestasis,
relationships. It is suggested that approximately 30%
peliosis hepatis (lacunar blood-filled cavities, which
of dilatative cardiomyopathy cases show a familial
come from central veins or from focal necrosis of
accumulation. In most cases the inheritance is auto-
hepatocytes) and proliferative changes of the liver struc-
somal dominant, rarely X-chromosomal or autosomal-
ture such as focal-nodular hyperplasia and liver aden-
recessive. Because the probability of manifestation and
omas (Nieschlag 1981; Rolf and Nieschlag 1998; Nakao
gene expression have a high variability, some other
et al. 2000).
predictive and environmental factors (e.g. viral infec-
A causal connection between AAS misuse and
tions or stress) can be responsible for the development
hepatocellular carcinoma has been described mostly
of the cardiomyopathy (Maisch et al. 2005).
in patients with other severe liver diseases (Gianni-
It has not been definitely clarified whether AAS
trapani et al. 2006; Clasing and Mller 2008). It has
cause arterial hypertension. In some cases AAS abuse
been hypothesized that AAS could play a key role in
led to a long-term (up to one year) elevation of blood
the development of steatosis hepatis, inhibiting the
pressure (Achar et al. 2010). Arterial hypertension
normal process of steroid biosynthesis and leading
induced by AAS abuse can persist up to one year after
to cholesterol storage (Turillazzi et al. 2011).
discontinuation of drug intake.
A slight increase of transaminases is mostly
High doses of AAS, especially in cases of simul-
reversible, and several weeks after discontinuation of
taneous consumption of several preparations, can
AAS normal ranges are achieved (Basaria 2010).
cause reduction of the HDL fraction of cholesterol
and increase of LDL cholesterol (Hartgens et al. 2004;
Kindermann 2006; Bonetti et al. 2008). These effects 25.2.5 Bones and muscles
on lipoprotein levels can be noted approximately two Anabolic androgenic steroids given in childhood or
months after the beginning of ASS use. Only several adolescence cause an acceleration of bone maturation
months after discontinuation of the administration in young athletes. At the end of puberty, activation of

538
Chapter 25: Sequelae of doping with anabolic steroids

the endochondrial bone formation leads to premature serum parameters of the malnourished patients with
closure of the growth zones with growth retardation chronic renal failure improved due to reduction of
(Rolf and Nieschlag 1998; Przkora et al. 2005; Sjqvist catabolism (Johnson 2000; see Chapter 17).
et al. 2008). Anabolic androgenic steroids including
testosterone support radial bone growth and distinct 25.2.7 Tumors
periost formation. This also explains the larger cross-
There is no indication that testosterone in replace-
section size of male compared to female bones
ment doses has any effect on tumor development or
(Vanderschueren et al. 2004; Lindberg et al. 2005;
growth, except in the prostate where it stimulates
see Chapter 7).
growth of an existing carcinoma (see Chapter 13).
Under acute intake of AAS rhabdomyolysis has
However, there are no reports about a relationship
been observed, with acute renal failure as a possible
between current or former AAS abuse and prostate
complication.
carcinoma.
Athletes often strain their musculoskeletal system
The most feared malignant disease after long-term
acutely and to an extreme extent over long periods,
AAS intake is hepatocellular carcinoma. The possible
resulting in a high incidence of complaints, injuries
cause of tumor development in the case of abuse of
and disorders in joints, tendons, bones and muscles.
17a-alkylated AAS is direct hepatotoxicity; in the case
These may become chronic so that the former athlete
of AAS underlying aromatization (endogenous testos-
suffers long after discontinuing high-performance
terone), a toxic effect of estrogens on hepatic tissue
sports and AAS abuse. There are, however, no
is discussed. It has been observed that human hepa-
appropriate investigations documenting a negative
tocellular carcinoma tissue has elevated aromatase
impact of AAS on the musculoskeletal system, and it
activity. Attempts to treat unresectable hepatocellular
is even suspected that AAS could possibly prevent
carcinoma with tamoxifen did not lead to positive
more severe damage.
results (Giannitrapani et al. 2006).

25.2.6 Kidneys 25.2.8 Behavior


Renal disorders have been described mostly after Headaches, sleeplessness, increased irritability and
long-term AAS use, and range from a slight increase depressive mood status were described following
of serum creatinine to acute renal failure as a compli- AAS abuse (Turillazzi et al. 2011). A study in women
cation of rhabdomyolysis. It has been hypothesized described complaints of depressive mood after discon-
that the interindividual differences concerning tinuation of AAS, persisting several weeks thereafter
the grade of side-effects depend on the genetically (Gruber and Pope 2000). Anabolic androgenic steroids
programmed function of the uridine diphosphate abusers have an affinity for developing alcohol or
glucuronosyltransferase (UGT) enzymes, which pro- opioid dependence (Basaria 2010). It has been sug-
vide glucuronidation of steroids, the first phase of gested that there are at least three etiological mechan-
the deactivation and elimination pathway of AAS isms of developing AAS dependence: body image
(Deshmukh et al. 2010). Histologically, a focal seg- disorders such as muscle dysmorphia; an experience
mental glomerulosclerosis with tubular atrophy and of dysphoria or depression after attempting to discon-
interstitial fibrosis can be found with long-term abuse tinue misuse, based on hypogonadotropic hypogonad-
(Turillazzi et al. 2011). Mild forms of renal dysfunc- ism; and possible hedonic effects of AAS (Kanayama
tion with elevation of serum creatinine, blood urine et al. 2010).
nitrogen and uric acid without sclerotic/fibrotic mor-
phological changes often return to normal ranges
after discontinuation of AAS (Turillazzi et al. 2011).
25.2.9 Causes of sudden death under
Conversely, testosterone and other AAS have been anabolic androgenic steroids
used for over 25 years for anemia treatment in patients In most cases of sudden cardiac death of young ath-
with chronic kidney insufficiency before erythropoietin letes, the cause was previously undiagnosed congeni-
became available for clinical use. However, the doses tal heart failure (Sullivan et al. 1998). As further
used were in the normal clinical range and far below possible causes the following are discussed: coronary
those in AAS abuse. The general condition and the spasms due to inhibition of NO release; premature

539
Chapter 25: Sequelae of doping with anabolic steroids

coronary arteriosclerosis due to increased atherogen- Considering the great number of teenage boys
esis; thrombotic coronary arterial occlusion due to using AAS, the question arises as to whether AAS
increased blood platelet aggregation and/or an application in boys around puberty may be harmful
increase of hematocrit and blood viscosity; as well as to spermatogenesis. Although systematic investiga-
direct cardiotoxic effects with impairment of mito- tions in pubertal AAS users are lacking, treatment of
chondria and myofibrils and associated destruction of over-tall boys with high doses of testosterone for
the myocytes and their replacement by fibrous tissue reduction of final height provides an analogy. Initially
(Dickerman et al. 1995; Sullivan et al. 1998; Fineschi it was suspected that this treatment would be harmful
et al. 2001; Kistler 2006). to the testes and leave permanent damage. However,
The above mentioned study describing autopsy when the proper control groups were co-investigated,
results and chemical-toxicological findings of 10 the incidence of subnormal semen parameters was the
young bodybuilders showed that 5 athletes died same in both groups (Lemcke et al. 1996; Hendriks
because of acute cardiac dysfunction due to myocar- et al. 2010), indicating that at this age the testes are
dial infarction (see Section 25.2.3). Further old infarc- not different from adult men in their capacity to
tions (n = 4) and chronic ischemic myocardial recover from suppression.
changes (n = 10) were found. Noteworthy was the fact In cases of high-dose intake of aromatizable
that AAS abusers had an affinity to consume other AAS, bilateral gynecomastia in men can develop with
substances. In 8 of 10 cases concomitant medication a prevalence of 2030% (OSullivan et al. 2000). Con-
was detected in blood and/or urine samples or in the current use of ER or aromatase inhibitors has been
hair. In three cases acute intoxication with opioids, applied to counteract this development. In cases of a
benzodiazepines, alcohol and Rohypnol (flunitrazepam) persistent, therapy refractory gynecomastia, a liposuc-
was the cause of death (Kistler 2006). tion with mastectomy may be required (Babigian and
Silverman 2001; Hartgens and Kuipers 2004).
After abrupt discontinuation of AAS abuse ath-
25.3 Specific side-effects in men letes can show temporary signs of hypogonadotropic
Due to negative feedback in the regulation of the hypogonadism, such as decreased libido, ED and
hypothalamic-pituitary-gonadal axis, AAS can cause depression (Basaria 2010).
a reversible suppression of spermatogenesis up to The single case of a former GDR weightlifter has
azoospermia (Knuth et al. 1989; Rolf and Nieschlag been reported who used Oral Turinabol at high doses
1998). As the spermatogenic tissue constitutes about (up to 20 tablets per day) from 18 to 23 years of age.
95% of the testes, its atrophy is followed by shrinkage He developed gynecomastia under the treatment
of the testes. After cessation of AAS intake spermato- and was, at the age of 32, operated on for a unilateral
genesis and testicular volumes recover within months. intratesticular leiomyosarcoma (Froehner et al. 1999).
During intake, the users may be infertile to various As these tumors are extremely rare and have been
degrees, often being unaware of the causal relation- described in hamsters after treatment with testoster-
ship. Proper diagnosis may be impeded by the fact one propionate and diethylstilbestrol (Kirkham and
that these men may not wish to admit abuse, neither Algard 1965), the authors suspected a causal relation-
to their physician nor to their partner, and an insist- ship between AAS abuse and the sarcoma. As this
ent exploration is required. Low LH, FSH and testos- remains the only reported case, the pathogenesis of
terone (in cases where testosterone is not used as the the tumor is unclear.
AAS) point to the suppressed pituitary-testicular axis
(Fronczak et al. 2011). Should spermatogenesis not
recover after cessation of use, a pre-existing fertility
25.4 Effects of anabolic androgenic
disorder is more likely than damage caused by the steroids in women
AAS. In order to hasten recovery, sometimes hCG is In females, dysmenorrhea, secondary amenorrhea
prescribed without any proof of effectiveness. It with anovulation and, as their consequence, infertility
should be mentioned here that the suppression of are the changes most often caused by AAS abuse.
the pituitary and spermatogenesis by testosterone is A large study with the aim of evaluating the side-
exploited in approaches to male hormonal contracep- effects of testosterone administration in therapeutic
tion (see Chapter 22). doses in women showed that there were no significant

540
Chapter 25: Sequelae of doping with anabolic steroids

differences concerning the frequency of cerebro- This dysarthria may become a problem for teachers,
vascular diseases, coronary heart disease, mamma actors and singers who are professionally dependent
carcinoma, deep venous thrombosis/lung embolism, on their voices. Such voice alterations are also
diabetes mellitus or acute hepatitis between women observed with endogenous elevation of testosterone
receiving testosterone therapy and the control group levels, e.g. CAH (Nygren et al. 2009) or in women
(Van Staa and Sprafka 2009; see also Chapter 23). sensitive to the androgenic action of some oral
Changes of the reproductive system due to sup- contraceptives. As changes of the voice are mostly
pression of the hypothalamic-pituitary-gonadal axis, irreversible, application of AAS or other steroids has
such as dysmenorrhea, secondary amenorrhea with to be suspended at the earliest sign of symptoms.
anovulation and reduction of breast size are revers-
ible. It can take weeks or months up to complete
recovery of the axis. In some cases it has been 25.4.3 The question of breast cancer
reported that after cessation of AAS administration The question about the effect of exogenous androgens
in women it can take up to two years until testoster- on the development of breast cancer has been dis-
one concentrations in serum dropped to normal levels cussed controversially in the scientific literature.
(Urman et al. 1991). Concerning possibly irreversible The lack of controlled studies contributes to the
side-effects of AAS use in women, such as clitoris uncertainties so that indirect evidence from other
hypertrophy, there is no documented experience. clinical situations has to be referred to.
Experience with long-term hormonal therapy in
25.4.1 Hirsutism transsexuals (female to male) aiming at virilization
(standard therapy: testosterone enanthate 250 mg
Hirsutism is the most frequent and reversible side-
im every second week or testosterone undecanoate
effect of AAS use in women (Braunstein 2007). The
1000 mg every 1012 weeks for two to three years
degree of increased facial or body hair growth
before surgical therapy, e.g. mastectomy, ovariectomy
depends on the dose and duration of AAS excess
and hysterectomy, and for years after that) shows
and can be described according to the hirsutism
no increased risk for breast cancer (Gooren et al.
score by FerrimanGallwey, established in 1961.
2008; Mueller and Gooren 2008). Since the 1970s,
Based on the intensity of hair growth in nine face/
when the first hormonal therapy of transsexuals was
body areas, hirsutism can be diagnosed as mild,
documented, only one clinical case has been reported;
moderate and severe (Ferriman and Gallwey 1961).
in this case a mamma carcinoma of the residual
In some cases it has been reported that after AAS
breast tissue developed 10 years after bilateral
administration in women it could take up to two
mastectomy and continuous testosterone therapy
years until testosterone concentrations in serum
(Burcombe et al. 2003).
dropped to normal levels and hirsutism disappeared
The polycystic ovary syndrome (PCOS) is charac-
(Urman et al. 1991).
terized by a significant increase of the testosterone
concentration in blood, and often serves as a model
25.4.2 Deepening of the voice for long-term testosterone exposure in women.
Deepening of the voice is part of the virilization Studies showed that the risk for breast cancer in
that AAS can cause in women. In contrast to acne, these women does not increase (Somboonporn and
hirsutism, alopecia, mammary atrophy and clitoral Davis 2004).
hypertrophy, deepening of the voice tends to be irre- Exogenous androgens are partially metabolized in
versible (Kindermann 2006). These effects of andro- the breast tissue to estrogens. However, not all syn-
gens in women have been described repeatedly (Bauer thetic androgens are subject to aromatization; e.g.
1973; Wirth 1979; Strauss et al. 1985; Baker 1999). tibolone and its metabolites cannot be aromatized
Lowering of the voice is caused by growth of the (de Gooyer et al. 2008). This also applies to the
larynx in girls and by thickening of the vocal chords metabolism of Oral Turinabol (chlordehydromethyl-
in women after puberty. The voice change can be so testosterone) unless taken in extremely high doses
pronounced that on the telephone women may be (see Section 25.3): the molecule is not aromatized,
mistaken for men. It is accompanied by hoarseness so that estrogenic side-effects become clinically not
which may intensify upon longer use of the voice. relevant (Kley 2004).

541
Chapter 25: Sequelae of doping with anabolic steroids

A large randomized study showed that postmeno- 25.5 Conclusion


pausal women who received estrogens exclusively did
As shown in this chapter, AAS abuse can result
not have an increased risk of mamma carcinoma, in
in minor and major sequelae, most reversible;
contrast to women who received an estrogen/gestagen
some irreversible and severe. Unfortunately, the
combination (Anderson et al. 2004). The age of the
negative and deserved image of AAS is often
patient and the duration of estrogen therapy are
applied to testosterone when used for a clinical
considered as risk factors for the development of
indication. Proper studies are lacking, and medical
breast cancer in women. Comparable results have also
knowledge of the field is predominantly based on
been shown in other studies (Magnusson et al. 1999;
case reports, so that causal relationships are often
Million Women Study Collaborators, 2003). How-
difficult to establish. Nevertheless, athletes, body-
ever, women who at the time point of the evaluation
builders and fitness studio customers appear to
received hormone replacement therapy (estrogen or
have only vague knowledge of the side-effects of
estrogen/gestagen preparations), in comparison to
AAS, which they often belittle in the light of the
women who had never taken hormonal drugs, had a
relatively few severe long-term problems in relation
higher risk for the development of breast cancer.
to the vast number of AAS abusers and the fact that
Women who in the past received hormonal therapy
AAS contribute significantly to performance and
did not have a higher risk for mamma carcinoma.
success. More education about the possible sequelae
It has also been shown that the additional admin-
of AAS abuse is mandatory to prevent negative
istration of testosterone during hormonal replace-
long-term effects. At the same time the medical
ment therapy in postmenopausal women (estrogen/
profession should pay more attention to the endo-
gestagen preparations) inhibited the proliferation of
crinology of physical activity and sports, and should
breast cells and thereby decreased the risk of mamma
research the question of why athletes have the
carcinoma (Hofling et al. 2007).
desire to use AAS, and whether they may need
In vitro, in animals and also in postmenopausal
some sort of androgen substitution to reach the
patients, androgens (e.g. testosterone, DHT) blocked
goals they set for themselves. Thereby medicine
proliferation of breast cells caused by estrogens and
could contribute to the problem beyond doping
expression of ER genes (Lapointe et al. 1999; Zhou
control and bring androgen abuse from the illicit
et al. 2000; And et al. 2002; Ortmann et al. 2002;
darkness to the light of knowledge, resulting in
Dimitrakakis et al. 2004). The antiproliferative and
healthy cooperation between athletics and medical
proapoptotic action of androgens is probably medi-
research.
ated through the AR, despite the potential of testos-
terone to metabolize to estrogens (Somboonporn and
Davis 2004). Before these interrelations were known, 25.6 Key messages
advanced stages of mamma carcinoma had even been
 Anabolic androgenic steroid effects and
treated with testosterone from the 1940s until the side-effects depend on dose, sex and weight of
1970s (Van Winkle 1949; Labhart 1978). The under- the athlete, duration of intake and combination
lying clinical experience was that testosterone inhibits with other drugs.
rather than supports a mamma carcinoma.  Due to the illicit nature of AAS abuse, controlled
Mutations in BRCA1 and BRCA2 genes (breast clinical trials on the short- and long-term
cancer genes) can exhibit a higher risk for the devel- undesired side-effects of AAS are lacking, so
opment of a mamma carcinoma. that a causal relationship between AAS abuse
In conclusion, there are no appropriate epidemi- and assumed sequelae is often difficult to
ological studies which clearly document or negate a establish.
causal connection between the administration of AAS  Anabolic androgenic steroids cause a temporary
activation of the coagulation system as well as
in young female athletes and the development of
fibrinolysis.
mamma carcinoma later in life. There is also no
 The most frequent AAS effect on the
accumulation of case reports which would argue for cardiovascular system is left ventricular
such a connection. Indirectly one can assume that use myocardial hypertrophy with impairment of
of AAS (e.g. Oral Turinabol) at young ages cannot be diastolic function.
causal for breast cancer.

542
Chapter 25: Sequelae of doping with anabolic steroids

 Effects on the liver (cholestasis, peliosis)  Anabolic androgenic steroid abuse does not
are associated with a chronic misuse of the contribute to the development of mammary
17a-alkylated AAS. carcinoma.
 Anabolic androgenic steroids do not increase the  Most AAS effects are reversible.
incidence of malignant tumors, with a possible  In unclear clinical situations the possibility
exception of 17a-alkylated AAS induced of AAS abuse should be taken into
hepatocellular carcinoma. consideration.

25.7 References of four women following hormonal


therapy with virilizing agents.
analysed to distinguish one disease
from the other? J Cardiovasc Med
Achar S, Rostamian A, Narayan SM J Voice 13:496507 (Hagerstown) 7:257266
(2010) Cardiac and metabolic
effects of anabolic-androgenic Bauer G (1973) Klinik der Clark BM, Schofield RS (2005) Dilated
steroid abuse on lipids, blood Stimmstrungen. In: Biesalski P cardiomyopathy and acute liver
pressure, left ventricular (ed) Phoniatrie. Thieme-Verlag, injury associated with combined use
dimensions, and rhythm. Am Stuttgart, pp 165172 of ephedra, gamma-
J Cardiol 106:893901 Basaria S (2010) Androgen abuse in hydroxybutyrate and anabolic
athletes: detection and steroids. Pharmacotherapy
Anderson GL, Limacher M, Assaf AR, 25:756761
Anderson GL, Limacher M, Assaf consequences. J Clin Endocrinol
AR, Bassford T, Beresford SA, Black Metab 95:15331543 Clasing D, Mller RK (2008)
H, Bonds D, Brunner R, Brzyski R, Berendonk M (1991) Doping- Dopingkontrolle, 5th edn.
Caan B, Chlebowski R, Curb D, Dokumente. Von der Forschung zum Sportverlag Strau, Cologne
Gass M, Hays J, Heiss G, Hendrix S, Betrug. Springer-Verlag, Berlin de Gooyer ME, Oppers-Tiemissen HM,
Howard BV, Hsia J, Hubbell A, Leysen D, Verheul HA,
Bhasin S, Storer TW, Berman N,
Jackson R, Johnson KC, Judd H, Kloosterboer HJ (2008) Tibolone is
Callegari C, Clevenger B, Phillips J,
Kotchen JM, Kuller L, LaCroix AZ, not converted by human aromatase
Bunnell TJ, Tricker R, Shirazi A,
Lane D, Langer RD, Lasser N, to 7alpha-ethynylestradiol
Casaburi R (1996) The effects of
Lewis CE, Manson J, Margolis K, (7alpha-MEE): analyses with
supraphysiologic doses of
Ockene J, OSullivan MJ, Phillips L, sensitive bioassays for estrogens and
testosterone on muscle size and
Prentice RL, Ritenbaugh C, androgens and with LC-MSMS.
strength in normal men. N Engl
Robbins J, Rossouw JE, Sarto G, Steroids 68:235243
J Med 335:17
Stefanick ML, Van Horn L, De Piccoli B, Giada F, Benettin A,
Wactawski-Wende J, Wallace R, Bonetti A, Tirelli F, Catapano A,
Dazzi D, Dei Cas A, Solito F, Sartori F, Piccolo E (1991) Anabolic
Wassertheil-Smoller S (2004) steroid use in body builders: an
Effects of conjugated equine Ceda G, Reverberi C, Monica C,
Pipitone S, Elia G, Spattini M, echocardiographic study of the left
estrogen in postmenopausal women ventricle morphology and function.
Magnati G (2008) Side effects of
with hysterectomy: the Womens
anabolic androgenic steroids abuse. Int J Sports Med 12:408412
Health Initiative randomized
controlled trial. JAMA 291:17011712 Int J Sports Med 29:679687 Deshmukh N, Petroczi A, Barker J,
Braunstein GD (2007) Safety of Szekely AD, Hussain I, Naughton
Aln M (1985) Androgenic steroid DP (2010) Potentially harmful
testosterone treatment in
effects on liver and red cells. Br advantage to athletes: a putative
postmenopausal women. Fertil Steril
J Sports Med 19:1520
88:116 connection between UGT2B17 gene
And S, De Amicis F, Rago V, Carpino deletion polymorphism and renal
Burcombe RJ, Makris A, Pittam M,
A, Maggiolini M, Panno ML, disorders with prolonged use of
Finer N (2003) Breast cancer after
Lanzino M (2002) Breast cancer: anabolic androgenic steroids. Subst
bilateral subcutaneous mastectomy
from estrogen to androgen receptor.
in a female-to-male transsexual. Abuse Treat Prev Pol 5:17
Mol Cell Endocrinol 193:121128
Breast 12:290293 Di Bello V, Giorgi D, Bianchi M,
Babigian A, Silverman RT (2001) Bertini A, Caputo MT, Valenti G,
Caso P, DAndrea A, Caso I, Severino
Management of gynecomastia due Furioso O, Alessandri L, Paterni M,
S, Calabr P, Allocca F, Mininni N,
to use of anabolic steroids in Giusti C (1999) Effects of anabolic-
Calabr R (2006) The athletes heart
bodybuilders. Plast Reconstr Chir androgenic steroids on weight-
and hypertrophic cardiomyopathy:
1:240242 lifters myocardium: an ultrasonic
two conditions which may be
Baker J (1999) A report on alterations misdiagnosed and coexistent. videodensitometric study. Med Sci
to the speaking and singing voices Which parameters should be Sports Exerc 31:514521

543
Chapter 25: Sequelae of doping with anabolic steroids

Dickerman RD, Schaller F, Prather I, after high dose doping with Oral- Kahn NN, Sinha AK, Spungen AM,
McConathy WJ (1995) Sudden Turinabol. Cancer 86:15711575 Bauman WA (2006) Effects of
cardiac death in a 20-year-old Fronczak CM, Kim ED, Barqawi AB oxandrolone, an anabolic steroid,
bodybuilder using anabolic steroids. (2011) The insults of illicit drug use on hemostasis. Am J Hematol
Cardiology 86:172173 on male fertility. J Androl Jul 28 81:95100
Dickerman RD, Schaller F, McConathy [Epub ahead of print] Kanayama G, Brower KJ, Wood RI,
WJ (1998) Left ventricular wall Gerber PA, Kukova G, Meller S, Hudson JI, Pope HG Jr (2010)
thickening does occur in elite power Neumann NJ, Homey B (2008) The Treatment of anabolic-androgenic
athletes with or without dire consequences of doping. Lancet steroid dependence: emerging
anabolic steroid use. Cardiology 372:656 evidence and its implications. Drug
90:145148 Alcohol Depend 109:613
Giannitrapani L, Soresi M, La Spada E,
Dimitrakakis C, Jones RA, Liu A, Cervello M, DAlessandro N, Karila TA, Karjalainen JE, Mntysaari
Bondy CA (2004) Breast cancer Montalto G (2006) Sex hormones MJ, Viitasalo MT, Seppl TA
incidence in postmenopausal and risk of liver tumor. Ann N Y (2003) Anabolic androgenic steroids
women using testosterone in Acad Sci 1089:228236 produce dose-dependant increase in
addition to usual hormone therapy. left ventricular mass in power
Gooren LJ, Giltay EJ, Bunk MC (2008)
Menopause 11:531535 athletes, and this effect is
Long-term treatment of
Evans NA (1997) Gym and tonic: a potentiated by concomitant use of
transsexuals with cross-sex
profile of 100 male steroid users. Br growth hormone. Int J Sports Med
hormones: extensive personal
J Sports Med 31:5458 24:337343
experience. J Clin Endocrinol Metab
Ferenchick GS, Hirokawa S, Mammen 93:1925 Kindermann W (2006)
EF, Schwartz KA (1995) Anabolic- Kardiovaskulre Nebenwirkungen
Gruber A, Pope HG Jr (2000)
androgenic steroid abuse in weight von anabol-androgenen Steroiden.
Psychiatric and medical effects of
lifters: evidence for activation of the Herz 31:566573
anabolic-androgenic steroid use in
hemostatic system. Am J Hematol women. Psychother Psychosom Kirkham H, Algard FT (1965)
49:282288 69:1926 Characteristic of an androgen/
Ferriman DM, Gallwey JD (1961) Hartgens F, Kuipers H (2004) estrogen dependent
Clinical assessment of body hair Effects of androgenic-anabolic leiomyosarcoma of the ductus
growth in women. J Clin Endocrinol steroids in athletes. Sport Med deferens of the Syrian hamster. I. In
21:14401447 34:513554 vivo. Cancer Res 25:141146
Fineschi V, Baroldi G, Monciotti F, Hartgens F, Rietjens G, Keizer HA, Kistler L (2006) Todesflle bei
Paglicci Reattelli L, Turillazi E Kuipers H, Wolffenbuttel BH Anabolikamissbrauch. Todesursache,
(2001) Anabolic steroid abuse and (2004) Effects of androgenic- Befunde und rechtsmedizinische
cardiac sudden death: a pathologic anabolic steroids on apolipoproteins Aspekte. Dissertation for Doctor of
study. Arch Pathol Lab Med and lipoprotein (a). Br J Sports Med medicine at the Medical Faculty of
125:253255 38:253259 the Ludwig-Maximilian University
in Munich
Fischer-Solms H (2009) Spitzensport: Hendriks AE, Boellaard WP, van
Doping in Ost und West. In: Casteren NJ, Romijn JC, de Jong Kley HK (2004) Anabole Steroide
Stiftung Haus der Geschichte der FH, Boot AM, Drop SL (2010) pharmakologische Wirkung der
Bundesrepublik Deutschland, Wir Fatherhood in tall men treated with Dopingmittel. In: Clasing D (ed)
gegen uns: Sport im geteilten high-dose sex steroids during Doping und seine Wirkstoffe.
Deutschland. Primus-Verlag, adolescence. J Clin Endocrinol Verbotene Arzneimittel im Sport.
Darmstadt, pp 111121 Metab 95:52335240 Spitta Verlag, Balingen, pp 6484
Franke WW, Berendonk B (1997) Hofling M, Hirschberg AL, Skoog L, Knuth UA, Maniera H, Nieschlag E
Hormonal doping and Tani E, Hgerstrm T, von Schoultz (1989) Anabolic steroids and semen
androgenization of athletes: a secret B (2007) Testosterone inhibits parameters in bodybuilders. Fertil
program of the German Democratic estrogen/progestogen-induced Steril 52:10411047
Republic government. Clin Chem breast cell proliferation in Labhart A (1978) Klinik der inneren
43:12621279 postmenopausal women. Sekretion, 3rd edn. Springer-Verlag,
Froehner M, Fischer R, Leike S, Menopause 14:183190 Heidelberg, pp 10391040
Hakenberg OW, Noack B, Wirth Johnson CA (2000) Use of androgens Lapointe J, Fournier A, Richard V,
MP (1999) Intratesticular in patients with renal failure. Semin Labrie C (1999) Androgens down-
leiomyosarcoma in a young man Dial 13:3639 regulate bcl-2 prooncogene

544
Chapter 25: Sequelae of doping with anabolic steroids

expression in ZR-75-1 human Reproductive Health and Sinner D, Bachmann M (2004) Anabole
breast cancer cells. Endocrinology Dysfunction, 3rd edn. Springer- Steroide. Das schwarze Buch.
140:416421 Verlag, Berlin, pp 437455 BMS Verlag, Gronau, pp 4749,
Nygren U, Sdersten M, Falhammar H, 561563
Lemcke B, Zentgraf J, Behre HM,
Kliesch S, Bramswig JH, Nieschlag E Thorn M, Hagenfeldt K, Sjqvist F, Garle M, Rane A (2008) Use
(1996) Long-term effects on Nordenskjld A (2009) Voice of doping agents, particularly
testicular function of high-dose characteristics in women with anabolic steroids, in sports and
testosterone treatment for congenital adrenal hyperplasia due society. Lancet 371:18721882
excessively tall stature. to 21-hydroxylase deficiency. Clin Somboonporn W, Davis SR (2004)
J Clin Endocrinol Metab 81: Endocrinol 70:1825 Testosterone effects on the breast:
296301 Ortmann J, Prifti S, Bohlmann MK, implications for testosterone
Lindberg MK, Vandenput L, Movrare Rehberger-Schneider S, Strowitzki therapy for women. Endocr Rev
Skrtic S, Vanderschueren D, T, Rabe T (2002) Testosterone and 5 25:374388
Boonen S, Bouillon R, Ohlsson C alpha-dihydrotestosterone inhibit in Steinigen J (2003) Zivilrechtliche
(2005) Androgens and the skeleton. vitro growth of human breast Aspekte des Dopings aus der Sicht des
Minerva Endocrinol 30:1525 cancer cell lines. Gynecol Endocrinol Spitzensportlers. Weiensee Verlag,
16:113120 Berlin
Magnusson C, Baron JA, Correia N,
Bergstrm R, Adami H-O, Persson I OSullivan AJ, Kennedy MC, Casey JH, Strauss RH, Liggett MT, Lanese RR
(1999) Breast cancer risk following Day RO, Corrigan B, Wodak AD (1985) Anabolic steroid use and
long-term oestrogen and (2000) Anabolic-androgenic perceived effects in ten weight-
oestsrogen-progestin-replacement steroids: medical assessment of trained women athletes. JAMA
therapy. Int J Cancer 81:339344 present, past and potential users. 253:28712873
Med J Aust 173:323327
Maisch B, Richter A, Sandmller A, Sullivan ML, Martinez CM, Gennis P,
Porting I, Pankuweit S (2005) Parkinson AB, Evans NA (2006) Gallagher EJ (1998) The cardiac
Inflammatory dilated Anabolic androgenic steroids: a toxicity of anabolic steroids. Prog
cardiomyopathy (DCMI). Herz survey of 500 users. Med Sci Sports Cardiovasc Dis 41:115
30:535544 Exerc 38:644651
Toorians A, Thomassen M, Zweegman
Million Women Study Collaborators Przkora R, Jeschke MG, Barrow RE, S, Magdeleyns E, Tans G, Gooren L,
(2003) Breast cancer and hormone- Suman OE, Meyer WJ, Finnerty CC, Rosing J (2003) Venous thrombosis
replacement therapy in the Million Sanford AP, Lee J, Chinkes DL, and changes of hemostatic variables
Women Study. The Lancet Mlcak RP, Herndon DN (2005) during cross-sex hormone
362:419427 Metabolic and hormonal changes of treatment in transsexual people.
severely burned children receiving J Clin Endocrinol Metab
Mueller A, Gooren L (2008) Hormone- long-term oxandrolone treatment. 88:57235729
related tumors in transsexuals Ann Surg 242:384389
receiving treatment with cross-sex Turillazzi E, Perilli G, Di Paolo M, Neri
hormones. Europ J Endocrinol Rolf C, Nieschlag E (1998) Potential M, Riezzo I, Fineschi V (2011)
159:197202 adverse effects of long-term Side effects of AAS abuse: an
testosterone therapy. Baillieres Clin overview. Mini Rev Med Chem
Nakao A, Sakagami K, Nakata Y, Endocrinol Metab 12:521534 11:374389
Komazawa K, Amimoto T,
Nakashima K, Isozaki H, Takakura Schollert PV, Bendixen PM (1993) Urman B, Pride SM, Yuen BH (1991)
N, Tanaka N (2000) Multiple Dilated cardiomyopathy in a user of Elevated serum testosterone,
hepatic adenomas caused by long- anabolic steroids. Ugeskr Laeger hirsutism, and virilism associated
term administration of androgenic 155:12171218 with combined androgen-estrogen
steroids for aplastic anemia in Sculthorpe N, Grace F, Jones P, Davies hormone replacement therapy.
association with familial B (2010) Evidence of altered cardiac Obstet Gynecol 77:595598
adenomatous polyposis. electrophysiology following Vanderschueren D, Vandenput L,
J Gastoenterol 35:557562 prolonged androgenic anabolic Boonen S, Lindenberg M, Bouillon
Nieschlag E (1981) Ist die Anwendung steroid use. Cardiovasc Toxicol R, Ohlsson C (2004) Androgens
von Methyltestosteron obsolet? 10:239243 and bone. Endocr Rev 25:389425
Dtsch Med Wochenschr Shapiro J, Christiana J, Frishman W Van Staa TP, Sprafka JM (2009) Study
106:11231125 (1999) Testosterone and other of adverse outcomes in women
Nieschlag E, Behre HM, Nieschlag S anabolic steroids as cardiovascular using testosterone therapy.
(eds) (2010) Andrology. Male drugs. Am J Ther 6:167174 Maturitas 62:7680

545
Chapter 25: Sequelae of doping with anabolic steroids

Van Winkle W Jr (1949) The action of and steroids. A short review. Acta estrogen receptor expression.
steroid hormones in mammary Dermatoven APA 16:117122 FASEB J 14:17251730
cancer. Radiology 53:530539 Winkler U (1996) Effects of androgens Zitzmann M, Nieschlag E (2007)
Vogt AM, Geyer H, Jahn L, Schnzer on haemostasis. Maturitas Androgen receptor gene CAG
W, Kbler W (2002) 24:147155 repeat length and body mass index
Cardiomyopathy associated with Wirth G (1979) Stimmstrungen. Dtsch modulate the safety of long-term
uncontrolled self medication of rzteverlag, Cologne intramuscular testosterone
anabolic steroids. Z Kardiol undecanoate therapy in
91:357362 Woodhouse LJ, Gupta N, Bhasin M, hypogonadal men. J Clin Endocrinol
Singh AB, Ross R, Phillips J, Bhasin Metab 92:38443853
Walker J, Adams B (2009) Cutaneous S (2004) Dose-dependent effects of
manifestations of anabolic testosterone on regional adipose Zitzmann M, Junker R, Kamischke A,
androgenic steroid use in tissue distribution in healthy young Nieschlag E (2002) Contraceptive
athletes. Int J Dermatol men. J Clin Endocrinol Metab steroids influence the hemostatic
48:10441048 89:718726 activation state in healthy men.
J Androl 23:503511
Wollina U, Pabst F, Schnlebe J, Abdel- Zhou J, Ng S, Adesanya-Famuiya O,
Naser MB, Konrad H, Gruner M, Anderson K, Bondy CA (2000) Zouboulis CC, Chen WC, Thornton
Haroske G, Klemm E, Schreiber G Testosterone inhibits estrogen- MJ, Qin K, Rosenfield R (2007)
(2007) Side-effects of topical induced mammary epithelial Sexual hormones in human skin.
androgenic and anabolic substances proliferation and suppresses Horm Metab Res 39:8595

546
Index

46,XX disorders of sexual mood effects, 1068 and bioavailable testosterone levels,
development, 36 sexual desire and response, 98106 3389
summary of evidence, 10910 and ejaculate volume, 336
Abaelard, Peter, 4 activin, 127 and erectile dysfunction, 2512
abdominal aortic aneurism, 342 acute coronary syndromes, 217 and fertility, 3367
abiraterone, 285 acute non-lymphocytic leukemia, 389 and hypogonadism, 323
acetylation of histones, 26 Addisons disease, 443, 444 and percentage spermatozoa with
acetyl-coenzyme A, 16 adipocytes normal morphology, 336
acne fat deposition, 23840 and sperm motility, 336
in women, 101 influence of DHT, 199 and total sperm output, 336
acne conglobata, 537 adipocytokines, 2389 androgen receptors, 339
acne vulgaris, 5367 adipogenic lineage, 254 androgen supplementation debate,
acromegaly, 62, 166 adipose tissue 337
activational effects of hormones, 88 hormones produced by, 2389 andropause, 336
activational effects of testosterone interactions with the HPT axis, aromatase activity, 340
(men), 908 23840 aromatization of testosterone,
aggression, 936 adrenal androgens 33940
comparison with women, 11014 age-related decline in women, 100 biotransformation in the tissues,
depression, 93 adrenal cortex 33940
effects on sleep, 967 fetal zone, 438 changes in coronary vasodilatation,
impact of aging, 90 adrenal glands 214
mood effects, 93 effects of over-stimulation, 107 effects in generally healthy men,
relevant information, 90 adrenal insufficiency, 440, 447 3367
sexual desire and response, 903 DHEA treatment, 4424 effects of co-morbidity on
summary of evidence, 978 adrenal steroidogenesis, 16 testosterone levels, 338
testosterone replacement in adrenalectomy, 254 effects of decline in testosterone
hypogonadism, 902 adrenarche, 37, 89, 438 levels, 323
activational effects of testosterone adrenopause, 438 effects of increasing fat mass, 340
(women), 98110 adult Leydig cells, 35 effects of obesity on testosterone
aggression and assertiveness, adverse effects of testosterone levels, 338
1089 strategies to avoid, 197200 effects of testosterone, 97
circulating androgens and sexuality, adverse events free testosterone levels, 3389
98100 testosterone therapy in older men, GnRH secretion, 3445
comparison with men, 11014 1956 gonadotropin levels, 3434
complexity of influences on age-related differences impact on effects of testosterone, 90
sexuality, 110 response to testosterone treatment, increase in SHBG binding capacity,
effects of aging, 100 193 345
effects of iatrogenic lowering of aggression increase of serum SHBG, 345
testosterone, 1003 effects of testosterone in men, 936 interindividual variability in effects,
effects on sexual problems in effects of testosterone in women, 3378
women, 1036 1089 Leydig cell function, 337
exogenous testosterone gender differences, 94 metabolic clearance rate for
administration and sexuality, aggressive personality testosterone, 337, 339
1036 and testosterone, 93 metabolism of androgens, 33940
extent of conversion to estrogen, Aggressive Provocation Questionnaire pathophysiology of declining
110 (APQ), 96 testosterone levels, 3435
individual variability of response, aging primary testicular changes, 3434
10910 altered neuroendocrine regulation, production of DHT, 33940
menopause, 100 3445 5a-reductase activity, 33940

547
Index

aging (cont.) genetic influences, 3401 prohormones of nortestosterone,


reduction in mean testicular influence of insulin levels, 341 521
volume, 343 interindividual variations, 340 prohormones of testosterone, 521
regulation of LH secretion, 3445 lifestyle factors, 342 supplements contaminated with
Sertoli cell function, 3367 metabolic factors, 341 prohormones, 523
serum testosterone levels, 3378 physiological factors, 3402 testosterone-to-epitestosterone
SHBG levels, 3389 serum testosterone levels in disease, ratio, 519
SHBG-bound testosterone, 339 3423 tetrahydrogestrinone (THG), 522
spermatogenesis, 3367 SHBG levels, 3412 use in sports, 1912, 221
testosterone conversion to Aging Male Symptom Score, 46 anabolic androgenic steroids side-
estradiol, 340 Albertus Magnus, 6 effects
testosterone production, 3378 albumin, 21, 60, 61, 273, 423 acne conglobata, 537
tissue levels of androgens, 33940 alcohol acne vulgaris, 5367
see also androgen therapy in effects on testosterone level, 342 amenorrhea in women, 541
elderly men. aldo-keto reductase (AKR), 24 anovulation in women, 541
aging and hypoandrogenism aldosterone, 442 atherosclerosis risk, 538
and depression, 3512 aldosterone antagonists, 219 athletes attitudes towards misuse,
and quality of life, 352 alendronate, 183 542
androgen status measurement, 352 Alzheimers disease, 351 azoospermia, 540
body composition, 3489 American Association of Clinical behavioral disturbances, 539
bone mineral density, 3501 Endocrinologists, 413 blood effects, 537
cardiovascular risk profile, 3478 American Society of Reproductive bones, 5389
clinical relevance, 34552 Medicine, 413 breast cancer risk, 5412
cognitive function, 351 amygdala, 89 cardiac arrhythmias, 537
effects on fat mass, 348 amygdaloid nuclei, 89 cardiac death risk in young
effects on mood, 3512 anabolic androgenic steroids, 9, 1912, athletes, 538
effects on muscle function, 3489 373, 461 causes of sudden death, 53940
effects on muscle mass, 3489 and cardiac disease, 208 deepening of voice in women, 541
erectile function, 3467 and liver disease, 208 depressive mood, 539
erythropoiesis, 351 and sudden cardiac death, 208 difficulty in establishing the cause,
hemoglobin levels, 351 anti-doping fight in sport, 5301 5356
long-term effects of androgen athlete doping program in the GDR, dilatative cardiomyopathy (DCM),
therapy, 348 518, 536 538
nocturnal penile tumescence (NPT), black-market products, 5201 dysmenorrhea in women, 541
346 designer steroids, 519, 5223 gynecomastia, 540
risk of falls, 349 development of synthetic steroids, heart and vessels, 5378
risk of fractures, 34951 51720 hepatocellular carcinoma, 538, 539
sarcopenia, 3489 discovery of, 1912 hirsutism, 5367, 541
senile osteoporosis, 34951 doping with Oral-Turinabol, 536 hypertension, 538
sexual function, 3467 extent of the doping problem, hypogonadotropic hypogonadism,
signs and symptoms of androgen 5356 540
deficiency, 3456 findings from anti-doping testing, increase in erythrocytes, 537
skeletal effects of testosterone, 520 increase in hematocrit, 537
34951 frequency of steroid misuse, 5201 increase in hemoglobin
aging and testosterone levels, 3403 history of misuse in sports, concentration, 537
and androgen receptor 51720 increased fibrinolysis, 537
polymorphisms, 341 illegal use and abuse, 9 intrahepatic cholestasis, 538
birth cohort effect, 341 misuse in bodybuilding, 520 intratesticular leiomyosarcoma, 540
body composition and adiposity, misuse in controlled competition kidneys, 539
341 sports, 520 liver, 538
drug side-effects, 343 misuse in fitness studios, 520 muscles, 5389
effects of alcohol, 342 misuse in non-controlled sports, need for more public education, 542
effects of diet, 342 5201 pathological myocardial
effects of medications, 343 prohibition by sports organizations, hypertrophy, 5378
effects of smoking, 342 51720 peliosis hepatis, 538
effects of stress, 342 prohormones, 519 rhabdomyolysis, 539
effects of thyroid hormone changes, prohormones of androgens, 5212 seborrhea, 5367
342 prohormones of skin effects, 5367
ethnic differences, 341 dihydrotestosterone, 521 specific effects in men, 540

548
Index

specific effects in women, 5402 androgen-induced hepatotoxicity, effects on reproductive function,


striae distensae, 5367 374 445
suppression of spermatogenesis, 540 androgen-induced polycythemia, 375 ethnic differences, 434
tumors, 539 androgen insensitivity syndrome gender differences in effects, 11112
anabolic androgenic steroids testing (AIS), 3842 hypotheses, 478
A and B sample testing, 5234 androgen receptor gene mutations, 41 influence on body tissues, 45
derivatization, 526 androgen-regulated genes, 42 influence on hair growth, 47
detection of endogenous anabolic clinical findings, 389 influence on sperm concentration,
androgenic steroids, 5289 functional characteristics of 45
detection of synthetic anabolic mutations, 412 influence on spermatogenesis, 445
androgenic steroids, 5268 genetics, 401 influence on testosterone therapy,
development of testing laboratory assessment, 3940 478
methodology, 524 somatic mosaicism, 41 Kennedy syndrome (XSBMA), 423
DHT doping, 528 androgen insufficiency syndrome mouse model of human sequence, 43
gas chromatographymass (AIS) pharmacogenetics, 478
spectrometry (GC-MS) method, effects on hair growth, 161 prostate development and
5245 androgen receptor (AR), 10, 15, 24, 88, malignancy, 434
high-resolution mass spectrometry 123, 215 psychological effects, 467
(HRMS), 528 alternative (non-nuclear) receptors, androgen receptor gene, 25
isotope ratio mass spectrometry 278 CAG repeat polymorphisms, 89
(IRMS), 5289 androgen-binding domain, 26 location, 40
metabolism and excretion, 525 cellular membrane receptors, 278 location and structure, 33
organization of doping tests, 5234 defects, 27 mutations, 41
pharmacokinetics, 525 distribution in the brain, 8990 role in androgenetic alopecia in
sample preparation, 5256 DNA-binding domain, 26 men, 165
sample taking and transportation, effects of aging, 339 role in organizational effects of
5234 effects of androgen administration, testosterone, 89
stages of analysis, 5245 27 somatic mosaicism, 41
testosterone-to-epitestosterone evolution of, 271 structure and function, 4038
ratio, 5289 expression on adipocytes, 240 androgen receptor GGC repeat
WADA-approved laboratories, 524 genomic pathway of action, 1356 polymorphisms, 25
anabolic hormones, 220 hinge region, 26 androgen receptor knock-out mice, 35,
Anabolic Steroid Control Act, 522 interaction with DNA, 26 36, 184, 212
anastrozole, 183, 211 mechanism of androgen action, androgen receptor modulators, 191
ancient Egypt, 2, 3 247 androgen receptor mutations, 10
androgen ablation therapy mediation of androgen action on androgen receptor pathophysiology
for metastatic prostate cancer, bone cells, 184 androgen insensitivity in humans,
2845 methylation/demethylation 3842
androgen binding protein (ABP), 15, enzymes, 26 effects of CAG repeat
21, 130 modulation of transcription activity, polymorphisms, 428
androgen-dependent reporter genes, 334 Kennedy syndrome (XSBMA), 423
412 non-genomic pathways of action, outlook, 489
androgen deprivation therapy, 10 136 prostate development and
cardiovascular risk, 209 N-terminal domain, 25 malignancy, 434
for prostate cancer, 209, 236, 238, phosphorylation, 26 androgen receptor polymorphisms, 309
240 regulation of action, 26 and testosterone levels in elderly
risk factor for metabolic syndrome, role in hair growth, 161 men, 341
238 selective androgen receptor and type 2 diabetes, 240
risk factor for type 2 diabetes, 238 modulators (SARMs), 27 see also androgen receptor CAG
androgen functions testicular androgen receptors, 1325 repeat polymorphisms.
development of external genitalia, transcriptional regulation, 40 androgen receptor TGG repeat
367 androgen receptor CAG repeat polymorphisms, 25
fetal prostate development, 356 polymorphisms, 25, 428, 240, androgen-regulated genes, 42
male puberty and adulthood, 378 341 androgen response elements (AREs),
masculinization of the fetus, 37 and benign prostatic hyperplasia 26, 40, 1356, 163
sexual differentiation of embryo and (BPH), 44 androgen sensitivity
fetus, 357 cardiovascular risk factors, 456 and type 2 diabetes, 240
target genes in fetal prostate effects of, 334 androgen sensitivity test, 40
development, 36 effects on bone tissue, 45 androgen status measurement, 352

549
Index

androgen suppression female pattern hair loss (FPHL), androstenone (16 ene-5a-androsten-
and spermiation failure, 134 166 3-one), 28
androgen therapy in elderly men, grading, 166 androsterone, 23
3528 incidence, 166 isolation of, 9
diagnosing androgen deficiency, androgenetic alopecia treatments, anemia due to bone marrow failure
3525 1678 androgen therapy, 3756
effects on the prostate, 3567 bimatoprost, 1678 anemia of end-stage renal failure
goals of androgen therapy, 3525 dutasteride, 168 androgen therapy, 3789
gynecomastia, 356 endocrine-based treatments, 168 anemias, 27
increase in hematocrit, 3556 finasteride, 168 anger
increase in hemoglobin levels, for eyelashes, 167 dimensions of, 95
3556 latanoprost, 1678 angina, 214
indications for treatment, 3525 minoxidil, 167 angina pectoris, 226
long-term benefitrisk ratio, 353 non-hormonal therapy, 1678 testosterone treatment studies,
modalities of androgen substitution, prostaglandin analogs, 1678 21718
357 5a-reductase inhibitors, 168 angioplasty, 214
potential benefits, 355 surgery, 167 angiotensin converting enzyme
potential risks, 3557 androgens, 22 inhibitors, 219, 222
sleep apnea, 356 effects on SHBG, 62 ankle-brachial index, 210
use of screening questionnaires, genomic effects, 247 Annibaldi, Domenico, 4
354 mode of action in the brain, anorchia, 39, 292
androgen therapy in non-gonadal 8990 anterior hypothalamic/preoptic area,
disease non-genomic effects, 278 88
approaches to androgen therapy, physiological effects, 15 antiandrogens, 285, 343
3723 role in spermatogenesis, 1308 effects in women, 101
body weight problems, 3945 androkinin (hypothetical male anti-arrhythmics, 219
bone disease, 3834 hormone), 8 anticonvulsants, 393
critical illness, 3846 andromedins, 277, 279, 281 antiepileptic drugs, 61
effects of androgen therapy, 373 andropause, 336 anti-Mullerian hormone (AMH), 35,
goals of androgen therapy, 373 androsetenedione 39
hematological disorders, 3757 intracrine activation, 442 anti-thymocyte globulin (ATG), 375
immune disease, 3868 androst-5-ene-3b,7b,17b-triol anxiety, 447
liver disease, 3745 (bAET), 440 aortic intima-media thickess, 210
malignant disease, 3889 androstan-3a-ol-17-one. Aphrodite (Venus)
muscular disorders, 3801 See androsterone birth of, 2
neurological disease, 3913 androstane aplastic anemia
renal disease, 37780 structure, 31011 androgen therapy, 3756
respiratory disease, 38991 androstane-3a,17b-diol, 16 apolipoprotein A1, 348
rheumatological disorders, 3813 5a-androstane-3a,17b-diol- apolipoprotein B, 347
steroid-induced osteoporosis, 384 glucuronide (ADG), 24, apolipoprotein E, 347
surgery rehabilitation, 3846 340, 440 apoptosis
trauma, 3846 5a-androstane-3,17-dione, 274 during brain development, 88
vascular disease, 3934 5a-androstane-3,17b-diol, 275 in rat Leydig cells, 21
androgenetic alopecia, 158, 1634 3a-androstanediol, 24 AR. See androgen receptor
androgenetic alopecia in men, 163, androsten-17a-ol-3-one. Arabic medicine, 6
1646 See testosterone areal bone density
and prostate cancer risk, 165 androstene-3,17-dione, 17 and testosterone deficiency, 1801
changes in the hair follicles, 164 androstenedione, 23, 26, 35, 99, 131, Aristotle, 2, 156
consequences of hair loss, 165 340, 438, 495 aromatase, 238
grading, 164 administration in clinical studies, action in men, 422
incidence, 1645 4467 activity in the brain, 89
link to myocardial infarction, 165 as a prohormone, 4412 in adipocytes, 238
pattern of balding, 164 effects of administration in women, aromatase activity, 234
role of, 1645 103 effects of aging, 340
role of androgens, 1656 in depressed women, 107 aromatase cytochrome P450 enzyme,
role of genes, 1656 in women, 100, 108 132
role of the androgen receptor gene, mechanisms of action, 4412 aromatase deficiency, 4234, 429, 432,
165 use as a food supplement, 447 433
androgenetic alopecia in women, 166 use by athletes, 447 caused by testosterone treatment, 24

550
Index

aromatase inhibitors, 238, 350, 432 Bartoli, Cecilia, 4 bone mass


aromatase knock-out mice, 23, 143, 426 Beck Depression Inventory (BDI), 93, monitoring testoterone therapy, 301
aromatization of testosterone to 101, 104 bone maturation
estradiol, 1434, 3501 bed nucleus of the stria terminalis role of estrogens in men,
arousal (BNST), 88, 89 4301
and testosterone level, 94, 967 behavioral effects. See activational bone metabolism
aryl-propionamides, 459, 461 effects of testosterone in elderly men, 34951
asthma benign prostatic hyperplasia (BPH), influence of estrogens in men,
androgen therapy, 391 27, 44, 270, 271, 272, 2812, 285, 42931
atheroma development 343, 416, 431 bone mineral density (BMD)
carotid artery atheroma, 210 and androgen therapy in elderly effect of DHEA treatment, 4489
coronary atheroma, 20910 men, 3567 effects of testosterone therapy for
peripheral arterial disease, 21011 SRD5A inhibitor treatment, 2812 women, 504
atherosclerosis, 224 Berthold, Arnold A., 6, 8 in elderly men, 3501
age-related effects, 2089 beta-adrenergic receptor blockers, 219, in hypogonadism, 41516
and low testosterone, 20911 222 influence of estrogens in men, 431
and testosterone levels, 2089 bicalutamide, 199, 285, 459 bone tissue
effect on testosterone levels, 342 bicyclic hydantoin, 461 effects of CAG repeat AR
effects of estrogens, 208 bimatoprost, 1678 polymorphisms, 45
fatty streak development in animal bioassay of testosterone, 78 bones
models, 21112 bioavailable testosterone, 778, 210, actions of estrogen, 23
plaque rupture, 215 211, 224, 226 actions of testosterone, 23
role of testosterone in animal effects of aging, 3389 osteoporosis, 23
models, 21112 birth cohort effect, 341 risk of fractures in elderly men,
atherosclerosis pathophysiology bloodtestis barrier, 124 34951
angina pectoris treatment, 21718 body composition side-effects of anabolic steroids,
effects of testosterone on serum effects of DHEA treatment, 448 5389
cytokine levels, 213 body mass index (BMI), 240, 341 boys on hemodialysis
endothelial adhesion molecule body weight problems androgen therapy, 380
expression, 21314 androgen therapy, 3945 growth of, 380
endothelial cell injury, 212 bodybuilding brain, 24
erythropoiesis, 216 misuse of anabolic steroids, 520 action of androgens and estrogens,
evidence for effects of low boldenone, 522 8990
testosterone, 21217 boldenone undecylenate, 520 distribution of androgen receptors,
fatty streaks, 216 bone disease 8990
features of advanced atheroma, androgen therapy, 3834 brain development
213 bone health gender differentation and
formation of fatty streaks, 21213 androgen receptor-mediated action testosterone, 889
hemostatic factors, 21516 on bone cells, 184 organizational effects of
plaque rupture, 213 dual actions of testosterone on testosterone, 889
role of LDL, 212 bone, 1834 white matter, 89
thrombotic process, 21516 effects of androgens on bone cells, brain imaging studies, 88
thrombus formation, 213 184 brain natriuretic peptide, 223
total and LDL cholesterol, 21617 osteoblast functions, 184 breast, 24
treatment studies, 21718 osteoclast functions, 184 breast cancer, 375, 439
vascular reactivity, 21415 role of estrogens, 178 contraindication for testosterone
atherosclerotic plaque, 212 role of testosterone, 1778 therapy, 303
athletes testosterone replacement in elderly Brief Index of Sexual Functioning for
use of androstenedione, 447 men, 182 Women (BISF-W), 106
see also anabolic androgenic testosterone replacement in Brief Psychiatric Rating Scale (BPRS),
steroids. hypogonadal men, 1812 95
attention deficit hyperactivity disorder see also osteoporosis. Brown-Squard, Charles E., 67
(ADHD) bone homeostasis buccal administration of testosterone,
and CAG repeat AR role of estradiol in men, 431 31415
polymorphisms, 46 bone marrow cells burn injury
autoimmune disease response to androgen therapy, androgen therapy, 3856
effects of testosterone therapy, 221 375 buserelin, 475
azoospermia, 25 bone marrow failure Buss and Perry Aggression
anabolic steroid side-effect, 540 androgen therapy, 3757 Questionnaire, 95, 96, 109

551
Index

BussDurkee Hostility Inventory carotid artery atherosclerosis cholesterol ester hydrolase, 18


(BDHI), 95, 96 effect on testosterone levels, 342 chromosome 2, 24
Butenandt, Adolf, 8, 9 carotid endarterectomy, 210 chromosome 5, 24
carotid-femoral pulse wave velocity, chronic anemia, 216
C-reactive protein (CRP), 245 211 chronic heart disease
cachectic patients, 216 Casodex, 285 anaboliccatabolic imbalance, 220
cachexia, 222 castor oil in testosterone preparations, cardiac cachexia, 220
effects of testosterone therapy, 221 3223 effects of testosterone on physical
CAG repeat polymorphisms. castration, 24, 254 function, 221
See androgen receptor CAG ancient Egypt, 3 effects of testosterone on skeletal
repeat polymorphisms as lawful punishment, 3 muscles, 221
CAG repeats (polyglutamine), 25 as revenge for seduction and effects on blood pressure, 220
calculated free testosterone, 78 adultery, 4 reduced cardiac output, 220
California Verbal Learning Test, 505 as revenge on enemies, 3 vasoconstriction, 220
Canadian Physicians guidelines, 413 cardiac effects, 221 chronic heart failure, 216, 21824, 226
cancers, 193 castrato singers, 4 androgen status, 220
cancers in women, 25 Chinese eunuch system, 23 baroreceptor sensitivity, 224
capon combs test, 8 effects on life expectancy, 4 brain natriuretic peptide marker,
cardiac cachexia, 220, 222 Greek mythology, 2 223
cardiac disease in the Islamic world, 3 breathlessness and fatigue, 219, 220,
and anabolic steroids, 208 medieval Scandinavia, 3 221
cardiac effects of testosterone Normans, 3 cachexia, 222
treatment, 221 of slaves, 23 cardiac cachexia, 222
cardiomyopathy, 223 of Uranos by Chronos, 2 cardiac effects of testosterone
cardiovascular disease prepubertal, 4 treatment, 221
and erectile dysfunction, 245, 252 range of supposed therapeutic uses, causes, 218
epidemiology, 2078 10 chronic anemia, 222
see also atherosclerosis; chronic self-mutilation for religious clinical features, 219
heart failure; coronary artery reasons, 3 see also orchidectomy. clinical trials of testosterone
disease. castration before puberty therapy, 2224
cardiovascular events effects on hair growth, 161 drug therapy, 219
and testosterone levels, 217 castrato singers, 4 effect of testosterone on exercise
testosterone therapy in older men, catabolic hormones, 220 capacity, 2234
1956 Catharers, 3 effect of testosterone on
cardiovascular events in women cell membrane hemodynamics, 222
effects of HRT, 208 effects of estradiol, 89 erythropoietic effects of
cardiovascular function in women effects of testosterone, 89 testosterone, 222
effects of testosterone therapy, cellular membrane androgen hematocrit, 222
504 receptors, 278 hemoglobin concentration, 222
cardiovascular mortality cellular therapy, 7 inflammation, 222
gender differences, 347 cetrorelix, 92 insulin resistance, 2212
cardiovascular risk cGMP, 252, 254, 255 metabolic syndrome, 220
and erectile function, 255 chaperones, 24, 26 mortality rate of severe heart failure,
and estrogen, 208 chemiluminescent assay (CLIA), 74 220
and metabolic syndrome, 235 chemotherapy, 343 non-cardiac effects of testosterone
and obesity, 235 Chinese eunuch system, 23 treatment, 221
and testosterone levels, 2089 Chinese medicine, 6 pathophysiology, 220
and testosterone therapy, 417 cholestasis of the liver, 312 prevalence, 219
and type 2 diabetes, 235 cholesterol, 16, 218 prognosis, 220
androgen deprivation therapy, 209, conversion to pregnenolone in reduced cardiac output, 222
238 mitochondria, 19 systemic vascular resistance,
CAG repeat androgen receptor conversion to testosterone, 1617 221, 222
polymorphisms, 456 effect of testosterone therapy, 245 systolic blood pressure, 224
insulin resistance, 2356 requirement by Leydig cells, 16 theoretical basis for testosterone
metabolic syndrome, 2367 role in atherogenesis, 21617 treatment, 221
carotid artery storage as lipid droplets, 16, 18 vasoconstriction, 221
intima-media thickness, 210, 211 transport to the mitochondria, chronic illness
carotid artery atheroma 1819 see also HDL cholesterol; effects of androgen therapy on
and low testosterone, 210 LDL cholesterol. physical function, 1934

552
Index

loss of muscle mass and function, congestive heart failure, 193, 356 dehydroepiandrosterone sulfate.
1934 constitutional delay of puberty See DHEAS
chronic obstructive pulmonary disease testosterone therapy, 302 Deiters, Otto, 1
(COPD), 193, 342, 343 contraception. See male hormonal delayed puberty in boys, 432
androgen therapy, 38990 contraception; oral contraception testosterone therapy, 302
effects of androgen therapy on (women) D5 pathway of steroid synthesis, 1617
physical function, 194 coronary angiography, 210 D4 pathway of steroid synthesis, 1617
chronic renal failure, 343 coronary artery disease, 212, 213 demasculinization
chronic respiratory failure, 375 and low testosterone, 209 in androgen receptor knock-out
chronic stable angina, 218 and sexual dysfunction, 251 mice, 36
chronic urticaria thrombotic process, 21516 in male knock-out mice, 37
androgen therapy, 383 coronary artery vasodilatation, 218 dementia, 439
Chronos coronary atheroma denosumab, 183
castration of Uranos, 2 and low testosterone, 20910 depression
cimetidine, 168 coronary atherosclerosis and CAG repeat AR
cingulate cortex, 91 effect on testosterone levels, 342 polymorphisms, 46
cingulate gyrus, 91 coronary disease, 222 and testosterone levels, 46, 96
circadian variation of serum coronary heart disease androgen therapy, 3923
testosterone, 340 and low testosterone, 2089 effect of DHEA treatment, 445
circannual variations in testosterone epidemiology, 2078 effects of testosterone in men, 93
levels, 340 gender differences in prevalence, effects of testosterone in women,
cirrhosis of the liver 2079 1068
androgen therapy, 374 coronary thrombosis, 215 effects of testosterone therapy,
claustrum, 91 corpus callosum, 88, 91 1967
clinically available forms of cortisol, 62, 438, 442 in elderly men, 3512
testosterone, 910 in depressed women, 107 Depression Scales, 46
clitoris couple relationship depressive disorders, 447
fetal development, 36 and testosterone levels, 261 dermal papilla, 1613
clobetasol propionate, 505 CREB transcription factor, 136 Derogatis Interview for Sexual
clomifene, 238 critical illness Function, 104
clomipramine, 107 androgen therapy, 3846 designer steroids, 519, 5223
cluster headaches effects on testosterone levels, 342 desogestrel, 4856
androgen therapy, 392 Crohns disease, 446 desoxymethyltestosterone (Madol,
coactivator proteins, 34 cryopreservation of sperm DMT), 522
cognitive function Spallanzani, 5 development
and testosterone levels in women, Cushing syndrome, 62, 166, 343 influence of testosterone, 97
500 Cushing, Harvey W., 7 DHEA, 17, 88, 131, 210, 347, 495
effects of androgen therapy, 3912 cyclic GMP. See cGMP adrenocortical steroid, 437
effects of testosterone therapy for cyclic quinolinones, 459 and atheroma formation, 211
women, 5045 cyclodextrins, 314 and well-being in women, 107
in elderly men, 351 cynomolgus monkeys, 89 effects on mood in women, 108
Colleoni, Bartolomeo, 4 CYP19 aromatase, 461 in women, 98, 100, 108
common marmoset (Callithrix CYP19 gene, 340, 350 mechanisms of action, 43941
jacchus), 128 cyproterone acetate, 101, 109, 155, use as anti-aging drug, 437
compensated hypogonadism, 344 168, 169, 4845, 537 DHEA deficiency
competitive androgen receptor cytochrome P450 aromatase, 89 approach to treatment, 449
antagonists, 285 cytochrome P450 oxido-reductase DHEA levels
complete androgen insensitivity deficiency (PORD), 432 age-related decline, 437, 438
syndrome (CAIS), 10, 24, 27, 33, cytochrome P450ssc, 19 as health status indicator, 439
35, 36, 389, 111 cytokine activation, 218 epidemiology of effects, 4389
see also androgen insensitivity cytokines, 220, 2445 gender differences in effects, 4389
syndrome (AIS). adipocytokines, 2389 genetic component, 438
conduct disorder (CD) effects of testosterone, 213 interindividual variability, 438
and CAG repeat AR pattern throughout the lifetime, 438
polymorphisms, 46 danazol, 61, 75 secretion and age, 438
congenital adrenal hyperplasia (CAH), de Graaf, Regnier, 5 DHEA mechanisms
19, 36 decapeptyl, 475 intracrinology concept, 440
congenital aromatase deficiency, deep vein thrombosis (DVT), 3934 ligand for a specific DHEA receptor,
4234 dehydroepiandrosterone. See DHEA 4401

553
Index

DHEA mechanisms (cont.) potency compared to testosterone, dickkopf 1 (DKK1), 164


neurosteroid, 440 23 dienogest, 485
precursor for active sex steroids, production, 23 diet
43940 production in women, 495 and testosterone levels in elderly
DHEA sulfotransferase (SULT2A1), role in fetal prostate development, men, 342
439 36 diethylstilbestrol (DES), 4278
DHEA therapeutic profile, 4479 synthesis by 5a-reductase, 24 digoxin, 219
body composition, 448 use in doping, 528 dihydrotestosterone. See DHT
central nervous system effects, 447 DHT gel 5a-dihydrotestosterone. See DHT
effect on mood, 447 as osteoporosis therapy, 183 dilatative cardiomyopathy (DCM), 538
effect on sexuality, 447 diabetes mellitus, 24, 42, 46, 218, 222 diminished ovarian reserve (DOR),
immune system effects, 449 and cardiovascular disease, 207 446
metabolic effects, 4478 and sexual dysfunction, 252 direct free testosterone assays, 767
skeletal effects, 4489 diabetes mellitus type 1, 254 Djungarian hamster, 139
skin effects, 449 diabetes mellitus type 2, 24, 226, 254, DMPA (depot MPA), 101, 47684
DHEA treatment 347 Doisy, Edmund A., 8
adrenal insufficiency, 4424 and androgen deprivation therapy, dominance
Crohns disease, 446 238 and testosterone level, 93, 94
diminished ovarian reserve (DOR), and androgen receptor doping
446 polymorphism, 240 use of anabolic steroids, 9
effect on depression, 445 and androgen receptor sensitivity, doping in sport. See anabolic
effect on fatigue, 4424 240 androgenic steroids; SARMs
effect on libido, 4424 and cardiovascular disease, 235 doping screens for athletes, 24
effect on mood, 4424, 445 and low testosterone, 2356 doping tests
effect on pubic hair growth, 443, 444 and metabolic syndrome, 2367 organization of, 5234
effect on well-being, 4424, 445 and testosterone deficiency, 416 Doppler ultrasound scanning, 210
elderly subjects, 4445 and testosterone level, 2378 drospirenone, 169
future directions, 44950 clinical implications of low drostanolone enanthate, 520
hypopituitarism, 4434 testosterone, 245 drostanolone propionate, 520
immunological disorders, 4456 effect of testosterone on body drug abuse
skin effects, 443, 444 composition, 242 and CAG repeat AR
systemic lupus erythematosus effect of testosterone on central polymorphisms, 46
(SLE), 4456 adiposity, 242 drug-related hirsutism, 166
ulcerative colitis, 446 effect of testosterone on cholesterol drugs
DHEAS, 88, 99, 226, 340, 347, 495 and lipoproteins, 245 effects on testosterone levels, 343
adrenocortical steroid, 437 effect of testosterone on Duchenne muscular dystrophy
and vitality in women, 107 dyslipidemia, 244 androgen therapy, 381
in women, 98, 100 effect of testosterone on ED, 245 dutasteride, 161, 168, 2812, 2856,
DHEAS levels effect of testosterone on 311
age-related decline, 437, 438 hyperglycemia, 2424 dyslipidemia
as health status indicator, 439 effect of testosterone on effect of testosterone therapy, 244
epidemiology of effects, 4389 hypertension, 244
gender differences in effects, 4389 effect of testosterone on echocardiography, 223, 224
genetic component, 438 inflammation, 2445 ED. See erectile dysfunction
interindividual variability, 438 effect of testosterone replacement, eflornithine hydrochloride, 168
pattern throughout the lifetime, 438 242 embryo
secretion and age, 438 effects in elderly men, 342 sexual differentiation, 357
DHT, 16, 23, 24, 127, 254 effects of testosterone on insulin endocriminology
5a-reduction of testosterone, 1312 resistance, 2401 description of organotherapy, 7
binding to AR in the prostate, 277 insulin resistance, 237, 242 endocrine function
cellular effects, 24 low testosterone as risk factor, and gonadal development, 345
effects of aging on production, 237 testosterone and the testes, 56
33940 potential benefits of testosterone Endocrine Society, 413
formation in the prostate, 273 replacement, 245 Endocrine Society of Australia, 413
glucuronoconjugated, 340 prevalence of hypogonadism, 235 endocrinology
in depressed women, 107 systolic blood pressure, 240 history of, 12
measurement of, 789 Diana/Artemis Ephesina fertility endoplasmic reticulum, 16
metabolism in the prostate, 274 cult, 2 endothelial cell adhesion molecule
metabolites, 24 diazepam, 103 expression, 21314

554
Index

endothelial glycocalyx, 21 evaluation of penile blood flow, 255 and cardiovascular risk, 208
endothelial nitric oxide synthase influence of sexual interest, 2545 estrogen in men
(eNOS), 4401 nocturnal erections, 2557 aromatase activity, 427
end-stage renal disease, 193, 211 nocturnal penile tumescence (NPT) estrogen-induced negative feedback,
androgen therapy, 3789 test, 257 100
effects of androgen therapy on penile color Doppler ultrasound estrogen receptor knock-out mice, 23
physical function, 194 (PCDU), 255 estrogen receptor a knock-out mice,
energy ROC curve analysis, 255 143, 426
effects of testosterone therapy, role of testosterone, 2535 estrogen receptor ab knock-out mice,
1967 sex-related erections, 257 426
ENERKI mice, 143 sleep-related erections (SREs), estrogen receptor b knock-out mice,
enuresis 2557 426
androgen therapy, 380 erectile tissues estrogen receptors (ERs), 25, 275
enzyme-linked immunosorbent assay excitatory tone, 91 estrogen receptor a (ERa), 132
(EIA/ELISA), 74 inhibitory tone, 91 estrogen receptor b (ERb), 132
epididymis, 24 erection physiology, 252 evolution of, 271
epilepsy, 393 corpora cavernosa, 252 in men, 4212, 423
equilibrium dialysis method, 756 detumescence, 253 polymorphisms, 45
ER. See estrogen receptors intracellular calcium, 252 estrogen replacement treatment in
erectile dysfunction (ED) nitric oxide (NO) formation, 252 men, 432, 433
and cardiovascular disease, 245, 252 nitric oxide synthase (NOS), 254 estrogen resistance, 432, 433
and hypogonadism, 25960 non-adrenergic, non-cholinergic estrogen response elements (EREs),
and metabolic disease, 252 (NANC) nerve endings, 252 132
and metabolic syndrome, 237 PDE5, 254 estrogen-to-testosterone ratio, 24
and testosterone deficiency, 237 psychogenic erection, 252 estrogens, 23
androgen level and erectile activity, reflexogenic erection, 252 and atherosclerosis, 208
2557 RhoA/ROCK pathway, 254 effects on SHBG, 62
biological dimension, 251 role of cGMP, 252, 254, 255 influence on testosterone effects,
definition, 251 role of noradrenaline, 252 234
effect of testosterone therapy, 245 role of phosphodiesterase (PDE5), mode of action in the brain, 8990
effects of sexual activity on 252 estrogens in men
testosterone levels, 2601 role of vasoactive intestinal peptide action on the hypothalamic-
effects of sexual inertia, 261 (VIP), 255 pituitary unit, 4234
effects of treatment on testosterone smooth muscle cells, 252 and gender identity, 4289
levels, 2601 venous leak, 254 and male infertility treatment, 432
evaluation of erectile funtion, erythrocytosis, 355 and sexual behavior, 429
2557 risk factor for testosterone therapy, and sexual orientation, 4289
impact on quality of life, 251 417 aromatase deficiency, 431, 432, 433
intrapsychic dimension, 251 erythropoiesis clinical implications, 432
meta-analyses of testosterone androgen therapy, 375 congenital aromatase deficiency,
therapy studies, 257 effects of testosterone, 216, 222 4234
organic dimension, 251 erythropoietin, 222, 375 congenital estrogen deficiency,
PDE5 inhibitor and testosterone esophageal cancer, 25 4234
combined therapy, 2579, 2612 estradiol, 16, 24, 26, 878 effects of aromatase deficiency, 429
prevalence, 2512 and aggression in women, 108 effects of inappropriate exposure,
prevalence of testosterone aromatization of testosterone to, 4278
deficiency, 25960 127, 1434, 3501 effects of prenatal exposure to
relational dimension, 251 in men, 422 diethylstilbestrol, 4278
relational domain, 261 production, 23 effects on human male
relationship with aging, 2512 production in elderly men, 340 reproduction, 427
erectile function role in bone homeostasis in men, effects on male reproduction, 4268
aging and hypoandrogenism, 3467 431 estradiol, 422
and cardiovascular risk, 255 SHBG binding, 22 estrogen biosynthesis, 4223
and MACE risk, 255 steroid feedback, 127 estrogen deficiency conditions, 432
and testosterone, 97 testicular, 132 estrogen receptors, 4212, 423
calcium sensitivity of smooth estradiol receptors, 89 estrogen replacement treatment,
muscle cells, 254 estradiol-to-testosterone ratio, 103 432, 433
cavernous peak systolic velocity estrogen, 22 estrogen resistance, 433
(PSV), 255 and aggression in women, 109 estrone, 422

555
Index

estrogens in men (cont.) Farinelli, Carlo, 4 FSH receptor, 19


historical development of fatal cardiovascular events fulvestrant, 212
knowledge, 4213 and testosterone levels, 217 functional brain imaging studies, 88
influence of circulating estrogens, fatigue
4234 effect of DHEA treatment, 4424 G-coupled receptor (LGR8), 21
influence on bone mineral density, fatigue/energy Galen, 5
431 effects of testosterone therapy, gambling (pathological)
influence on glucose metabolism, 1967 and CAG repeat AR
4312 fatty acid metabolite receptors, 25 polymorphisms, 46
influence on lipid metabolism, 432 fatty streak development, 211, 21213 gangliosides-associated testosterone
influence on male sexuality, 4289 female pattern hair loss (FPHL), 166 transport, 23
knock-out models of estrogen female sexual dysfunction (FSD) gas chromatographymass
deficiency, 4267 testosterone therapy, 5004 spectrometry (GC-MS), 634
male rodent reproduction models, fertile-eunuch syndrome, 10 derivatization, 63
4267 fertility gender differences
regulation of gonadotropin and aging, 3367 action of androgens and estrogens
feedback, 4234 constant fertility in human males, in the brain, 8990
role in bone maturation, 4301 272 aggression, 94
role in bone metabolism, 42931 fetal development of the testis, 16 and testosterone, 11012
role in boys before puberty, 433 fetal Leydig cells, 16, 35 AR distribution in the brain, 8990
role in sperm production and fetus cardiovascular mortality, 347
quality, 427 development of external genitalia, differentiation of the brain, 889
role of estradiol in bone 367 effects of DHEA/DHEAS, 4389
homeostasis, 431 development of the genital tubercle, osteoporosis, 1778
testosterone-to-estradiol ratio, 427 367 prevalence of coronary heart
therapeutic implications, 432 masculinization, 367 disease, 2079
threshold level for estradiol effect, sexual differentiation, 357 see also activational effects of
431 fibrinogen, 347, 348 testosterone.
estrone, 8, 23 fibrinolysis, 218 gender differences and testosterone
in men, 422 finasteride, 36, 155, 161, 168, 169, 274, amount of individual variability,
ethinyl estradiol, 62, 168 2812, 2856, 311, 343, 347, 351 110
ethnic differences fitness studios by-product hypothesis, 11314
CAG repeat AR polymorphisms, misuse of anabolic steroids, 520 desensitization hypothesis, 11213
434 flow-mediated brachial artery effects of androgen receptor gene
prostate development and vasodilatation, 215 polymorphisms, 11112
malignancy, 434 flow-mediated brachial reactivity, 211 explanatory hypotheses, 11214
testosterone levels in elderly men, fluoroimmunoassay (FIA), 74 mechanisms of androgen
341 fluoxymesterone, 313, 520 production, 111
etonogestrel, 4856 flutamide, 169, 212 relationship between mood and
eunuchoidal body proportions, 299 follicle stimulating hormone. See FSH sexuality, 11011
eunuchs, 254 follistatin, 127, 199 sensitivity to testosterone, 110
evolution of male accessory glands, forskolin, 26 threshold effect, 111
2689 fractures gender identity, 4289
excitatory tone in erectile tissues, 91 risk in elderly men, 34951 genetic factors
exercise testing in angina studies, frailty testosterone levels in elderly men,
21718 androgen therapy, 380 3401
external quality assessment (EQA), Free Androgen Index (FAI), 99 genital skin, 24
812 free radicals, 212 genital tubercle
externalizing behaviors free testosterone, 21, 273 development in the fetus, 367
and CAG repeat AR effects of aging, 3389 genomic effects of androgens, 247
polymorphisms, 46 measurement methods, 758 genomic pathway of androgen
extracellular matrix factors, 163 frontal lobe, 91 receptor action, 1356
eyelashes frustration tolerance German Democratic Republic
treatment for hypotrichosis, 167 and testosterone level, 93 secret athlete doping program, 518,
FSH (follicle stimulating hormone), 536
Fallopian tubes 21, 378, 39, 126 gestagens, 47686
development of, 35 action on Sertoli cells, 127 gestation
falls and spermatogenesis, 13840 development of the testis, 16
risk in elderly men, 349 feedback regulation, 127 GGC repeats (polyglycine), 25

556
Index

glucocorticoid receptors, 25, 40, 271 diagnostic criteria for melanocyte stem cells, 158
glucocorticoids, 21, 343, 442 hypogonadism, 414 paracrine factors in
glucose metabolism different approaches to guidelines, mesenchymeepithelial
role of estrogens in men, 4312 413 interactions, 1634
glucose tolerance erectile dysfunction (ED), 415 structure, 1578
impaired, 234 erythrocytosis risk, 417 hair follicle transplant surgery, 160
glucuronoconjugated DHT, 340 evidence-based versus expert hair growth
Glut-4, 241 opinion criteria, 413 after puberty, 157, 15960
glutamine (CAG) repeats, 25 hormone threshold level, 414 axillary hair, 159, 161
glycine (GGC) repeats, 25 impaired libido, 41415 balding inhibitors, 1634
glycolysis, 241 lower urinary tract obstructive balding scalp, 162
GnRH (gonadotropin-releasing symptoms, 416 beard, 158, 159, 161, 162, 163
hormone), 27, 37, 126, 127 metabolic syndrome, 416 beard hair color, 163
secretion in elderly men, 3445 monitoring patients on testosterone beard hair medulla, 163
GnRH agonists, 192, 193, 4745 treatment, 417 before birth, 159
GnRH analogs, 92, 130, 144, 343, mood, 416 before puberty, 15960
4746 osteoporosis, 41516 effects of androgens, 15961
GnRH antagonists, 92, 127, 131, 139, potential risks from testosterone effects of castration before puberty,
311, 4756, 496 therapy, 41617 161
GnRH deficiency, 128 principal published guidelines, 408 evidence for the role of androgens,
GnRH immunization, 129, 140 prostate cancer contraindication for 1601
GnRH receptor, 19 treatment, 416 eyelashes, 167
gonadal development reference to a specific age group, 413 in androgen insensitivity syndrome,
and endocrine function, 345 review committees, 41718 161
gonadal dysgenesis, 39 screening for androgen deficiency in androgen insufficiency
gonadarche, 37 syndrome, 414 syndrome, 161
gonadotropin-releasing hormone. selection of guideline committee in children, 154
See GnRH members, 413 influence of CAG repeat AR
gonadotropins, 16, 126, 127 specificity of diagnostic tests, 413 polymorphisms, 47
levels in elderly men, 3434 strength of the evidence base, intermediate hairs, 159
neuroendocrine control, 3445 41718 lanugo hairs, 159
gonads symptoms required for male pattern baldness, 161
tumor risk in AIS, 39 hypogonadism diagnosis, 414 mesenchymalepithelial
Graafian follicles, 5 treatment contraindications, 41617 interactions, 159
Greco-Roman period, 2 treatment options, 417 molecular mechanisms, 159
growth factors, 21 untreated sleep apnea risk factor, 417 production of hairs, 1578
production by dermal papillae, 163 well-being, 416 pubic hair, 159
growth hormone, 62, 345 gynecomastia, 24, 38, 42, 294, 2989, regulation by androgens, 1546
excess, 61 356, 486 requirement for DHT in men, 161
role in testosterone action on anabolic steroid side-effect, 540 role of androgen receptors, 161
muscles, 199 role of growth hormone, 161
guidelines for testosterone deficiency hair role of the dermal papilla, 1613
benign prostatic hyperplasia risk, functions of hair, 1567 seasonal variations, 1589
416 male sexual hair pattern, 298 terminal hair, 159, 161
bone mineral loss, 41516 sexual hair growth in CAIS, 38 variations within and between races,
cardiovascular risk from treatment, sexual hair growth in PAIS, 38 159
417 social and sexual functions, 154 vellus hair, 159
clinical manifestations of structure, 1578 hair growth cycle, 1589
hypogonadism, 41416 hair follicle, 24 hair growth disorders
clinically relevant testosterone level, capacity for regeneration, 1589 androgen-dependent conditions,
414 dermal papilla, 157, 158 1647
cognition, 416 epithelial stem cells, 158 androgenetic alopecia, 158, 1634
decreased muscle mass and epithelialmesenchymal androgenetic alopecia in men,
strength, 415 interactions, 158 1646
definition of a guideline, 408 extracellular matrix components, androgenetic alopecia in women,
definition of hypogonadism, 413 163 166
depression, 416 hair growth cycle, 1589 androgenetic alopecia treatments,
diabetes mellitus, 416 mechanism of androgen action, 1678
diagnosis of hypogonadism, 413 1614 balding inhibitors, 1634

557
Index

hair growth disorders (cont.) eflornithine hydrochloride, 168 effects of androgen therapy on
hirsutism, 166 endocrine-based treatments, 1689 physical function, 1934
hirsutism treatments, 1689 non-hormonal approaches, 168 effects of testosterone therapy, 221
treatment of androgen-dependent histone acetyltransferase (HAT), 277 purpose of androgen therapy, 222, 386
conditions, 167 histone demethylases, 25 HOMA-IR (Homeostatic Model
hair pigmentation, 163 histones Assessment of Insulin
Hamilton Depression Rating Scale acetylation, 26 Resistance), 240
(HDRS), 95 methylation, 26 hormone
Hamm, Johan, 5, 155 phosphorylation, 26 origin of the term, 12
Harvey, William, 5 ubiquitination, 26 hormone replacement therapy (HRT),
hCG (human chorionic history of testosterone and the testes, 45, 216, 496
gonadotropin), 1920, 144, 343 111 and cardiovascular events in
in marmosets and humans, 128 anabolic steroids, 9 women, 208
hCG receptors, 20 ancient Egypt, 2, 3 effects of testosterone
hCG therapy androkinin, 8 administration, 1045
in hypogonadotropic Arabic medicine, 6 HPG. See hypothalamic-pituitary-
hypogonadism, 12930 capon combs test, 8 gonadal axis
HDL cholesterol, 46, 210, 212, 21617, castration, 24, 10 HPT. See hypothalamic-pituitary-
237, 240, 244, 347, 348, 432 cellular therapy, 7 testicular axis
headache chemical synthesis of testosterone, Huggins, Charles, 10
androgen therapy, 392 810 human chorionic gonadotropin.
heat shock protein 90 (HSP 90), 26 Chinese eunuch system, 23 See hCG
height Chinese medicine, 6 Hunter, John, 56
testosterone therapy to reduce final clinically available forms of hydantoin derivatives, 4612
height, 3023 testosterone, 910 hydroxyflutamide, 459
hematocrit early descriptions of hypogonadism 17a-hydroxylase-17,20-lyase
effects of testosterone, 216 syndromes, 10 deficiency, 432
hematological disorders first descriptions of sperm, 5 17-hydroxypregnenolone, 17
androgen therapy, 3757 first descriptions of testicular 17-hydroxyprogesterone, 23
hemochromatosis, 343 morphology, 5 17a-hydroxyprogesterone, 17
hemodialysis Greco-Roman period, 2 17-hydroxyprogesterone capronate, 476
androgen therapy for boys on, 380 intramuscular testosterone hydroxysteroid dehydrogenase, 24
hemoglobin A1c (HbA1c), 2424 undecanoate, 10 11b-hydroxysteroid dehydrogenase, 21
hemoglobin concentration Islamic world, 3 17b-hydroxysteroid dehydrogenase,
effects of testosterone treatment, isolation of androsterone, 9 439, 440
217 isolation of testosterone, 9 17b-hydroxysteroid dehydrogenase
hepatitis LoeweVoss test, 8 deficiency, 38
androgen therapy, 374 medieval Scandinavia, 3 3b-hydroxysteroid dehydrogenase,
hepatocellular carcinoma, 538, 539 the Netherlands, 5 439, 440
hepatocytes Normans, 3 hypercholesterolemia
actions of sex steroids, 22 origins of endocrinology, 12 and cardiovascular disease, 207
hepatotoxicity pharmacokinetic studies, 9 hyperglycemia, 237
androgen-induced, 374 power through polyorchidism, 45 effect of testosterone on, 2424
17a-methyltestosterone, 312 proof of endocrine function, 56 hypergonadotropic hypogonadism,
hepcidin, 375 prostate cancer, 10 912, 181
hereditary angioedema ring structure of steroids, 8 hyperlipidemia
androgen therapy, 381 rise of steroid biochemistry, 8 and sexual dysfunction, 252
Hershberger assay, 461 Romans, 6 hyperprolactinemia, 166, 343, 498
high-altitude residence, 375 testicular organotherapy, 67 hypertension, 237, 356
high-density lipoprotein. See HDL testis transplantations, 78 and cardiovascular disease, 207
cholesterol testosterone immunoassays, 9 and sexual dysfunction, 251
high-resolution mass spectrometry transdermal testosterone gel, 10 effect of testosterone therapy, 244
(HRMS), 528 transdermal testosterone patches, 9 hyperthyroidism, 61, 341
hijras (India), 3 twentieth century, 810 hypertriglyceridemia, 237
hip fracture in elderly men, 350 HIV/AIDS, 193 hypoactive sexual desire disorder
hippocampus, 89 androgen therapy for wasting, (DSM-IV), 105
hirsutism, 101, 166, 541 3867 hypoactive sexual desire disorder
evaluation approach, 166 androgen therapy in patients (HSDD) in women
hirsutism treatments, 1689 without wasting, 3878 testosterone therapy, 5004

558
Index

hypogonadal (hpg) mice, 128, 132, male imprinting, 272 and ovarian androgen production,
137, 143 hypothalamo-hypophyseal-testicular 100
hypogonadal men axis, 1267 and SHBG, 2401
testosterone replacement, 902 hypothalamus, 90, 239 effects in elderly men, 342
hypogonadal osteoporosis, 180 hypothyroidism, 61, 62, 343 effects of testosterone therapy for
Hypogonadal-Obesity-Adipocytokine hypoxia, 375 women, 504
Hypothesis, 238 hysterectomy in chronic heart failure, 2212
hypogonadism, 2, 6, 27 effects of, 496 mechanisms of testosterone action,
and erectile dysfunction, 25960 effects on sexuality, 1023 2401
and estrogen deficiency, 432 hysterectomy and bilateral insulin sensitivity, 348
and obesity, 236 ovariectomy effect of testosterone replacement,
cardiovascular risk, 255 effects of testosterone 2412
choice of testosterone therapy, administration, 1056, 181 mechanisms of testosterone action,
2947 2401
classification, 294 idiopathic male infertility insulin-like growth factor (IGF), 226
effects of testosterone treatment on ineffectiveness of testosterone insulin-like growth factor 1 (IGF-1),
muscle mass, 1923 treatment, 303 163, 164, 168, 281, 341, 345
effects on sleep, 967 IGF. See insulin-like growth factor role in muscle growth and
intramuscular testosterone IL. See interleukin differentiation, 199
undecanoate, 10 immature Leydig cells, 16 insulin-like factor 3 (INSL3), 21, 35
level for initiation of testosterone immune disease interleukin 1 (IL-1), 239
therapy, 297 androgen therapy, 3868 interleukin 1b (IL-1b), 213, 239, 2445
non-hormonal anti-osteoporotic immune system interleukin 6 (IL-6), 239, 241, 2445
drugs, 183 effects of DHEA treatment, 449 interleukins, 222
range of applications of testosterone immunoglobulin, 220 internal quality control
therapy, 2924 immunological disorders measurement of testosterone, 80
skeletal effects of testosterone DHEA treatment, 4456 International Index of Erectile Function
replacement, 1812 importin-a, 26 (IIEF) questionnaire, 259
symptoms of testosterone importin-b, 26 International Index of Erectile
deficiency, 294 impotence. See erectile dysfunction (ED) Function (IIEF-5), 257
symptom-specific thresholds for impulsivity interstitial nuclei of the anterior
treatment, 297 and testosterone level, 93 hypothalamus (INAH 14), 88
testosterone treatment for angina, incremental shuttle walk test, 223 Interviewer Ratings of Sexual Function
21718 Indian hedgehog (Ihh), 252 (IRSF), 101
transdermal testosterone gel, 10 infectious disease susceptibility intracrine activation of
type 2 diabetes risk factor, effects of testosterone, 28 androstenedione, 442
2378 see also guidelines for inferior frontal gyrus, 91 intracrinology concept, 440
testosterone deficiency. infertility (male) intrahepatic cholestasis, 538
hypogonadism syndromes ineffectiveness of testosterone intramuscular administration of
early descriptions, 10 treatment, 303 testosterone, 31523
hypogonadism treatment inflammation, 218 intratesticular leiomyosarcoma, 540
effects of CAG repeat AR and obesity, 235 intratesticular testosterone, 128, 1302
polymorphisms, 478 effect of testosterone therapy, differences between animals and
hypogonadotropic hypogonadism, 2445 humans, 130
912, 239, 261 effects of testosterone, 222 suppression, 134
after anabolic steroid abuse, 540 in chronic heart failure, 216, 222 irritability
hCG therapy, 12930 role in prostate carcinogenesis, 282 and aggression, 93
hypophysectomy, 254 inflammatory cytokines, 212 and testosterone level, 93, 96
hypopituitarism, 2, 255, 440, 4434 inhibin B, 39, 127, 336 premenstrual syndrome (PMS), 106
in women, 104, 498, 504 inhibitory tone in erectile tissues, 91 isolated hypogonadotropic
hypospadias, 37 inotropes, 220 hypogonadism, 292, 294
hypotestosteronemia, 209 insula, 91 isotope ratio mass spectrometry
hypothalamic-pituitary-gonadal insulin levels, 341 (IRMS), 5289
(HPG) axis, 16, 48, 343 insulin receptor substrate 1 (IRS-1),
hypothalamic-pituitary-testicular 241 Japanese men
(HPT) axis, 22, 1267, 131, 273, insulin resistance, 234, 209, 218, 222, cardiovascular risk, 255
423 2356, 237
interactions with adipose tissue, and mitochondrial dysfunction, 241 Kallmann syndrome, 10, 292
23840 and obesity, 239 Karolinska Scales of Personality, 46

559
Index

Kennedy syndrome (XSBMA), 25, 34, LH receptor, 1920 major cardiovascular events
423, 44, 45, 46, 238 activating mutations, 128 and testosterone levels, 217
ketoconazole, 285 comparison between human and maldescended testes, 292
11-ketotestosterone, 28 marmoset, 128 male accessory glands, 2689
kisspeptin, 23940 inactivating mutations, 128 male adulthood
Klinefelter syndrome, 2, 8, 10, 181, LH receptor knock-out mice, 19, 128, androgen functions, 378
221, 238, 240, 255, 292, 297, 300 131 male climacteric, 10
LHRH (luteinizing hormone-releasing male contraception, 27
lactation, 98 hormone) super-agonists, 284, condom, 471
sexual interest during, 100 285 desirable features, 471
testosterone level variations, 100 life expectancy existing methods, 471
Laqueur, Ernst, 8, 9 effects of testosterone, 4 new approaches, 471
latanoprost, 1678 lifestyle reasons for, 4701
late-onset hypogonadism, 292, 345, effects on testosterone levels, 342 vasectomy, 471
413 lipid droplet storage of cholesterol, 16, male hormonal contraception, 47, 92,
LDL cholesterol, 16, 218, 244, 245, 18 130, 131, 132, 134, 138, 139, 144,
347, 432 lipid metabolism 318
role in atherogenesis, 21617 role of estrogens in men, 432 acceptability, 487
role in atherosclerosis lipoprotein (a), 245 adverse events, 486
pathophysiology, 212 lipoprotein lipase, 240 depression side-effect, 486
LDL receptor knock-out mice, 211 lipoproteins effects on liver function, 486
Leeuwenhoek, Antoni A., 2, 5 effect of testosterone therapy, 245 efficacy studies, 4723
leptin, 239, 240, 242, 244, 341, 345 liquid chromatographytandem mass future outlook, 487
leuprorelin, 92, 93, 96, 343 spectrometry (LC-MS/MS), 63, goal of azoospermia, 4712
levonorgestrel, 484 645 goal of oligozoospermia, 4712
Leydig cell function derivatization, 64 gynecomastia side-effect, 486
effects of aging, 337, 343 liver, 24 MENT (7a-methyl-19-
effects of drugs, 343 first-pass metabolism of nortestosterone), 474
effects of endocrine diseases, 343 testosterone, 310, 314 new approaches, 471
neuroendocrine control, 344 hepatotoxic effects of 17a- 19-nortestosterone, 474
Leydig cells, 5, 15, 35, 37, 123, 132, methyltestosterone, 312 19-nortestosterone plus DMPA,
133, 239, 292 metabolism of testosterone, 31011 47684
cholesterol homeostasis, 16 liver cirrhosis, 310, 342 principle, 4712
development from perivascular and liver disease, 343 side-effects, 4867
peritubular mesenchymal-like androgen therapy, 3745 testosterone alone, 4724
cells, 16 liver disorders testosterone buciclate, 473
fetal development of the testis, 16 androgen induced, 374 testosterone enanthate, 4723
in the testicular intersitium, 15 liver function testosterone pellets, 474
influence of LH, 16 during testosterone therapy, 301 testosterone plus cyproterone
interactions with Sertoli cells, 21 liver tumors, 312 acetate, 4845
ontogeny, 16 locus coeruleus, 91 testosterone plus desogestrel, 4856
proliferation in the adult testis, 16 Loewe, S., 8 testosterone plus dienogest, 485
regulation by factors other than LH, LoeweVoss test, 8 testosterone plus DMPA, 47684
21 loop diuretics, 219 testosterone plus etonogestrel,
steroidogenesis, 130 low-density lipoprotein. See LDL 4856
stimulation of androgen secretion, cholesterol testosterone plus GnRH agonists,
12930 lower urinary tract obstructive 4745
testosterone production, 1516 symptoms (LUTS), 416 testosterone plus GnRH analogs,
types of, 16 luteinizing hormone. See LH 4746
use of cholesterol, 16 Lydston, G. Frank, 7 testosterone plus GnRH
Leydig, Franz, 5 lymph circulation, 313 antagonists, 4756
LH (luteinizing hormone), 16, 37, 39, lymphatic circulation of the testes, 22 testosterone plus levonorgestrel,
126, 260, 261 lysine-specific demethylase 1 (LSD1), 484
influence on Leydig cell 26 testosterone plus 19-norethisterone,
development, 16 486
levels in AIS, 39 MAIS (minimal androgen insensitivity testosterone plus progestins, 47686
regulation of testosterone syndrome), 39 testosterone undecanoate (castor
biosynthesis, 1920 major adverse cardiovascular events oil), 4734
rise at menopause, 100 (MACE) risk, 255 testosterone undecanoate (oral), 473

560
Index

testosterone undecanoate (tea seed internal quality control, 80 effect of testosterone on


oil), 473 liquid chromatographytandem dyslipidemia, 244
weight gain side-effect, 486 mass spectrometry (LC-MS/MS), effect of testosterone on ED, 245
male imprinting of the HPT axis, 272 63, 645 effect of testosterone on
male infertility treatment, 432 mass spectrometry methods, hypertension, 244
male pattern baldness 6373 effect of testosterone on
influence of CAG repeat AR non-radioactive assays, 745 inflammation, 2445
polymorphisms, 47 quality control, 745, 7982 effect of testosterone on insulin
see also androgenetic alopecia. radioimmunoassay, 734 resistance, 2401
male puberty and adulthood reference ranges, 62 effect of testosterone replacement, 242
androgen functions, 378 salivary testosterone, 77 effects in elderly men, 342
male sexual development, 345 ultrafiltration, 76 insulin resistance, 237, 242
malignant disease validation of methodology, 7980 low testosterone as risk factor, 237
androgen therapy, 3889 validation of methods, 745 potential benefits of testosterone
mammalian evolution medial pre-optic area, 89 replacement, 245
dangers of reproduction, 2712 medications prevalence of hypogonadism, 235
seasonal breeding, 2712 effects on testosterone levels, 343 risk of developing diabetes, 2367
selection of testosterone as master Medvei, Victor C., 1 metandienone, 518, 520, 524
reproductive regulator, 2702 megalin, 21 metenolone acetate, 520
mammary gland development, 38 megestrol acetate, 476 metenolone enanthate, 520
mammillary nuclei, 89, 90 menopause, 98, 100 metformin, 169
manic behavior effects on mood, 107 methandriol dipropionate, 520, 538
associated with testosterone level, 96 surgical, 1056 7a-methyl-19-nortestosterone.
MAP-kinase pathway, 27 menstrual cycle, 98 See MENT
Marrian, Guy, 8 influence on mood, 106 methylation of histones, 26
marsupials, 25 testosterone level variations, 99 methyltestosterone, 96, 105, 520, 538
masculinization of the fetus, 367 MENT (7a-methyl-19- 17a-methyltestosterone, 6, 9, 312, 314
mass spectrometry nortestosterone), 91, 474 hepatotoxic side-effects, 312
measurement of testosterone, 6373 MENT implants, 484 micropenis
McCormick, Katherine, 487 mental retardation, 255 testosterone therapy, 303
M-CSF (macrophage-colony mesonephric ducts, 35 microphallus
stimulating factor), 184, 185 mestanolone, 518 testosterone therapy, 303
MDV-3100, 285 mesterolone, 303, 313, 520 mifepristone, 75
measurement of DHT, 789 metabolic clearance rate for mineralocorticoid receptors, 25, 40,
free and protein-bound forms, testosterone, 337, 339 271
602 metabolic disease mineralocorticoids, 442
measurement of testosterone, 6278 and erectile dysfunction, 252 minimal androgen insensitivity
assessment of free testosterone, metabolic effects of DHEA treatment, syndrome. 33 See MAIS
758 4478 minoxidil, 155, 167
automatic multianalyzers, 74 metabolic risk factors in men, 24 miRNAs, 136
bioassay, 78 metabolic syndrome, 45, 254, 347 mitochondria
bioavailable testosterone, 778 and androgen deprivation therapy, conversion of cholesterol to
calculated free testosterone, 78 238 pregnenolone, 19
challenges, 60 and androgen receptor transport of cholesterol to, 1819
chemiluminescent assay (CLIA), 74 polymorphism, 240 mitochondrial dysfunction
choice of kit, 7980 and cardiovascular disease, 235 and insulin resistance, 241
clinical applications, 62 and erectile dysfunction, 237 mitogens
comparison of different methods, and low testosterone, 2356 insulin-like growth factor (IGF-1),
6573 and testosterone deficiency, 416 163
direct free testosterone assays, 767 cardiovascular risk factors, 2367 Modified Mania Rating Scale
EIA/ELISA, 74 clinical implications of low (MMRS), 95
equilibrium dialysis, 756 testosterone, 245 17a-mono-oxygenase inhibitors, 285
external quality assessment (EQA), definitions, 2367 monotremes, 25
812 effect of testosterone on body mood
fluoroimmunoassay (FIA), 74 composition, 242 and testosterone level, 95
free and protein-bound forms, effect of testosterone on central effects of bilateral ovariectomy in
602 adiposity, 242 women, 1078
gas chromatographymass effect of testosterone on cholesterol effects of DHEA adminstration in
spectrometry (GC-MS), 634 and lipoproteins, 245 women, 108

561
Index

mood (cont.) role of growth hormone, 199 nocturnal penile tumescence (NPT),
effects of DHEA treatment, 4424 role of IGF-1, 199 346
effects of lowering of testosterone in strategies for selectivity in androgen effects of testosterone, 91
women, 107 therapy, 197200 nocturnal penile tumescence (NPT)
effects of oral contraceptives in testosterone as a function- test, 257
women, 107 promoting therapy, 200 non-genomic effects of androgens, 278
effects of testosterone in men, 93 testosterone therapy in healthy non-genomic pathways of androgen
effects of testosterone in women, older men, 1946 receptor action, 136
1068 testosterone treatment in non-SHBG-bound testosterone, 778
effects of testosterone therapy, hypogonadism, 1923 norboletone, 522
1967 muscle performance norethandrolone, 524
effects of testosterone therapy for effects of testosterone on reaction norethindrone, 476
women, 504 time, 197 19-norethisterone, 476, 486
influence of the menstrual cycle in influence of testosterone, 192 norethisterone enanthate, 23
women, 106 muscle wasting normal (eugonadal) men
menopausal symptoms, 107 androgen therapy, 3845 effects of increasing testosterone,
premenstrual syndrome (PMS), 106 muscular disorders 923
relationship to sexuality in women, androgen therapy, 3801 experimental reduction of
107 muscular dystrophies testosterone, 923
mood disorders androgen therapy, 3801 Normans, 3
and CAG repeat AR musculo-skeletal system Norplant, 101
polymorphisms, 46 side-effects of anabolic steroids, nortestosterone, 520, 524
effect of DHEA treatment, 445 5389 19-nortestosterone, 474, 484
Moreschi, Alessandro, 4 myeloproliferative disorders nortestosterone decanoate, 520
Morris, J.M., 10 androgen therapy, 3767 nortestosterone esters, 520
mortality predictive marker myocardial infarction, 213, 215, 217, novelty-seeking behavior
low testosterone level, 2246 238, 245, 355 and CAG repeat AR
mouse model of human AR CAG and sexual dysfunction, 251 polymorphisms, 46
repeats, 43 effects on testosterone levels, 342 nuclear androgen receptor, 212, 215
MPA (medroxyprogesterone acetate), link to androgenetic alopecia in nuclear receptor superfamily
476 men, 165 evolution of, 271
MRI brain scanning, 89 myocyte-specific androgen receptor nuclear receptors, 247
Mullerian ducts, 35 knock-out mice, 199 Nussbaum, Moritz, 8
Mullerian inhibiting substance (MIS), 35 myogenic lineage, 254 nutritional supplements
Multi-Dimensional Anger Inventory myotonic dystrophy contamination with prohormones,
(MAI), 95 effects of androgen therapy, 3801 523
multiple sclerosis, 393
muscle nafarelin, 475 obesity, 218
anabolic effects of androgens, 1912 NalGlu (GnRH antagonist), 92 and cardiovascular disease, 235
muscle function nandrolone, 351, 461 and hypogonadotropic
in elderly men, 3489 nandrolone decanoate, 194, 223 hypogonadism, 342
muscle mass nasal administration of testosterone, and low testosterone, 2356, 237
effects of lowering testosterone 315 androgen therapy, 3945
levels, 192
in elderly men, 3489

Nebido , 10, 218
negative mood
central obesity and insulin
resistance, 240
influence of CAG repeat AR and testosterone level, 96 effect of testosterone replacement,
polymorphisms, 45 neocortex, 89 242
influence of testosterone, 192 neonatal androgen secretion, 1278 effect of weight loss on testosterone
muscle mass and strength neonatal Leydig cells, 16 levels, 236
avoiding negative effects of the Netherlands hormones produced by adipose
testosterone, 197200 early anatomical discoveries, 5 tissue, 2389
dose-response relationship for neuroleptic drugs, 343 insulin resistance, 242
testosterone, 193 neurological disease risk factor for metabolic syndrome,
effects of androgen therapy in androgen therapy, 3913 235
chronic ilnesses, 1934 neurosteroids, 88 risk factor for type 2 diabetes, 235
effects of testosterone replacement, nexin-1, 1634 obesity measures
1923 nifedipine, 218 and testosterone levels, 192
mechanism of testosterone actions, nitric oxide synthase (NOS), 252, 254, obstructive sleep apnea
1979 346 androgen therapy, 3901

562
Index


Okasa , 6
older men
secondary osteoporosis, 180
osteoporosis (women)
and sexual dysfunction, 252
peripheral vascular venous disease,
effects of testosterone on sleep, 97 postmenopausal, 179 3934
oligozoospermia, 25 osteoporosis therapy peritubular cells, 21, 23, 126
Olweus Multifaceted Aggression anastrozole, 183 peritubular myoid cells, 35, 123, 124,
Inventory, 108 aromatase inhibitor, 183 132, 133, 135
opera DHT gel, 183 personality traits
castrato singers, 4 non-aromatizable androgen, 183 influence of CAG repeat length, 46
opiates non-hormonal drugs for Pzard, A., 8
effects on testosterone levels, 343 hypogonadal men, 183 PGE1, 255
oppositional defiant disorder (ODD) raloxifene, 182 pharmacogenetics
and CAG repeat AR SARMs, 182 CAG repeat AR polymorphisms,
polymorphisms, 46 SERMs, 182 478
oral administration of testosterone, testosterone replacement in elderly pharmacokinetic studies of
31013 men, 182 testosterone preparations, 9
oral contraceptive use (women), 99, testosterone replacement in pharmacokinetics of testosterone
103, 208, 216, 496 hypogonadal men, 1812 esters, 31516
effects on mood, 1001, 107 osteoprotegerin (OPG), 184 Philipp Magnanimous, Count of
effects on sexuality, 1001 ovarian androgens, 100 Hesse, 5
effects on testosterone levels, 1001 ovariectomy (bilateral) phorbol esters, 26
for hirsutism, 1689 effects of hormone replacement, phosphodiesterase inhibitors, 6
Oral-Turinabol, 518, 520, 536, 540, 1023 phosphodiesterase type 5 (PDE5), 254
541 effects of testosterone phosphodiesterase type 5 inhibitor
orbitofrontal cortex, 91 administration, 1056 (PDE5i), 346, 415
orchidectomy, 240 effects on mood, 1078 and testosterone combined therapy,
and prostate cancer, 10 effects on sexuality, 1023 2579
health effects, 209 effects on well-being, 1023 phosphorylation
see also castration. ovary, 24 androgen receptors, 26
orchitis, 292 function of the interstitial cells, 100 histones, 26
organizational effects of hormones oxandrolone, 461, 520, 538 physical function
brain development, 889 oxymetholone, 520 androgen therapy for chronically ill
organotherapy, 67 patients, 1934
orphan nuclear receptors, 25 P450 aromatase, 439, 440 effects of testosterone therapy in
osteoblasts, 184, 241 P450c17 enzyme, 439 older men, 1946
osteocalcin, 182 paramesonephric ducts, 35 in older men, 192
osteoclasts, 184 parathyroid hormone (PTH), 183 influence of testosterone, 192
osteocytes, 184 paraventricular nuclei, 90 potential use of testosterone
osteopenia, 27 Parkinsons disease, 393 therapy, 200
diagnosis from bone density, 1801 Parsifal, 3 pineal-hypophysis-pituitary system,
osteoporosis, 23, 27, 294, 301 partial androgen insensitivity 158
and testosterone in women, syndrome (PAIS), 10, 24, 33, pioglitazone, 169
499500 389 pituitary, 123, 126
functions of osteoblasts and see also androgen insensitivity expression of hGC in marmosets,
osteoclasts, 184 syndrome (AIS). 128
gender differences, 1778 Partner Aggression Questionnaire gonadotropin inhibition, 144
in hypogonadism, 41516 (AQ-P), 96 hypothalamo-hypophyseal-
postmenopausal, 183 peliosis hepatis, 312, 374, 538 testicular axis, 1267
preclinical rodent models, 1834 penile color Doppler ultrasound pituitary insufficiency, 292, 343
role of testosterone, 1778 (PCDU), 255 pituitary tumors, 343
steroid induced, 384 penile growth and development pituitary-gonadal function, 341
osteoporosis (men) role of testosterone, 2523 placenta, 24
and areal bone density, 1801 penis plaque rupture, 213
and testosterone deficiency, fetal development, 36 plasminogen activator inhibitor 1
17881 testosterone therapy for micropenis, (PAI-1), 215, 239
diagnosis from bone density, 1801 303 plasminogen activator type 1, 347
hypogonadal osteoporosis, 180 pentoxifylline, 220 Point Subtraction Aggression
idiopathic male osteoporosis, 17980 peripheral arterial disease Paradigm (PSAP), 95
risk factors for senile osteoporosis, and low testosterone, 21011 polycystic ovary syndrome (PCOS),
1789 peripheral vascular disease 166, 169, 541

563
Index

polycythemia, 300, 355 evolution of male accessory glands, ethnic differences, 434
androgen induced, 375 2689 high-grade prostatic intraepithelial
polyglutamine polymorphisms, 25 gene transcription and growth, 24 neoplasia (HGPIN), 2824
polyorchidism interactive stem cell units, 2779 intratumoral steroid synthesis,
power associated with, 45 intermediate cells, 279 285
polyproline (TGG repeats), 25 maintenance of steady-state size, preventative role for SRD5A
positive mood 279 inhibitors, 2856
and testosterone level, 96 mechanism of androgen action, proliferative inflammatory atrophy,
prairie dog (Cynomys ludovicanus), 139 2779 283
prefrontal cortex, 90 mesenchymal stem cells, 281 role of androgens in carcinogenesis,
pregnancy, 61, 62 monitoring testosterone therapy 2824
pregnandiol, 8 effects, 301 role of chronic and acute
pregnane, 16 neuroendocrine cells, 279 inflammation, 282
pregnenolone, 16, 23 organization of epithelial and treatment side-effects in aging men,
conversion of cholesterol to, 19 stromal cell units, 27981 2867
Premarin, 105 regenerative capacity, 279 use of supraphysiological levels of
premenstrual syndrome (PMS), 106 role of testosterone in development testosterone, 286
prepubertal castration, 4 and growth, 2723 prostate cancer cells, 28
primary spermatocytes, 124 role of testosterone in maintenance, prostate cancer risk, 165
primates 273 and androgenetic alopecia in men,
SHBG, 61 secretory products in humans, 270 165
testis development, 16 secretory-luminal cells, 27981 and CAG repeat AR
Profile of Female Sexual Function self-renewal of stem cells, 279 polymorphisms, 434
(PFSF), 99 side-effects of testosterone therapy, prostate development
Profile of Mood States (POMS), 96 2867 and CAG repeat AR
progenitor Leydig cells, 16 steady-state self-renewing tissue in polymorphisms, 434
progestagen receptors, 271 adults, 273 role of androgens in the fetus, 356
progesterone, 17, 23, 26, 89 stromalepithelial cell interactions, prostate ontogeny, 2778
progesterone receptors, 23, 25, 28, 40 2779 paracrine interactions, 2778
progestins, 28, 168, 47686 testosterone as a prohormone, 277 prostate-specific antigen (PSA), 26, 27,
progestogens, 92 testosterone binding to AR, 277 270
programmed cell death testosterone metabolism in, 2735 proteohormone, 21
during brain development, 88 transit-amplifying (TA) cells, 279 Provera, 105
prohormones transition zone, 281 psychological effects
androstenedione, 4412 variations between animal species, CAG repeat AR polymorphisms,
contamination in nutritional 26970 see also benign prostatic 467
supplements, 523 hyperplasia (BPH). psychosexual function
of anabolic androgens, 5212 prostate cancer, 25, 43, 165, 269, 270, and testosterone level, 95
of dihydrotestosterone, 521 271, 272 PTM-ARKO mice, 135
of nortestosterone, 521 and androgen therapy in elderly puberty, 21
of testosterone, 521 men, 3567 androgen functions, 378
testosterone as, 277 and orchidectomy, 10 effects of androgen receptor defects,
testosterone for estrogen, 421 and testosterone treatment, 10 27
use in doping in sports, 519 androgen ablation therapy for hair growth after, 15960
prolactin, 97 metastatic cancer, 2845 hair growth before, 15960
prolactinoma, 2, 343 androgen deprivation therapy, 209, testosterone therapy for delayed
proline (TGG) repeats, 25 236, 238, 240 puberty, 302
prostaglandin D synthase, 35 AR conversion to oncogene, 284 timing of, 889
prostate, 23, 24, 271 autonomous autocrine AR signaling pubic hair growth
age-related effects, 2867 pathway, 2834 effect of DHEA treatment, 443, 444
AR integration of androgen bipolar androgen therapy, 286 pulsatile testosterone production, 20
signaling, 2757 cardiovascular risk from treatment,
autoregulation of androgen 209 quality control
metabolism, 277 castration-resistant prostate cancer, immunoassay methods, 745
development of the urogenital sinus, 2845, 286 measurement of testosterone, 7982
2778 cell of origin, 2824 quality of life
DHT binding to AR, 277 contraindication for testosterone effects of testosterone therapy,
dual agonist/antagonist effects of therapy, 303, 416 1967
androgen, 273 effects of hormonal treatment, 343 quinolinones, 461

564
Index

radioimmunoassay for plasma rheumatological disorders for masculinization of the fetus,


testosterone, 734 androgen therapy, 3813 367
raloxifene, 182 RhoA/ROCK pathway, 254 septic shock, 440
RANKL (receptor activator of nuclear risedronate, 183 SERMs (selective estrogen receptor
factor-kB ligand), 184, 185 RNA synthesis, 25 modulators), 182
rat Leydig cells rodents Sertoli cell AR knock-out (SCARKO)
apoptosis, 21 testis development, 16 mice, 133, 134, 136
Rathus Assertiveness Schedule, 96 RoDH, 24 Sertoli-cell-only syndrome, 10, 292
reaction time rosiglitazone, 169 Sertoli cells, 5, 16, 21, 35, 127
effects of testosterone, 197 Rynearson, E.H., 7 action of testosterone on, 126
reactive oxygen species (ROS), 185 effects of aging, 3367
rectal administration of testosterone, Sabbatsberg Sexual Self-Rating Scale, interactions with Leydig cells, 21
315 104 role in spermatogenesis, 124, 126,
5a-reductase, 440, 461 salivary testosterone measurement, 77 1325
expression in the skin, 62 Sanger, Margaret, 487 role in testis development, 35
synthesis of DHT, 24 SARMs (selective androgen receptor stimulation by FSH, 44
274 see also SRD5A isoenzymes modulators), 27, 177, 182, 373 tight junctions, 135
5a-reductase activity characteristics, 459 Sertoli, Enrico, 5
effects of aging, 340 conformational hypothesis of serum amyloid A, 239
in the brain, 89 action, 4635 sex hormone-binding globulin.
5a-reductase deficiency, 38 discovery programs, 45960 See SHBG
5a-reductase genes, 165 early phase I and II trials, 466 sex hormones
5a-reductase inhibitors, 168, 311, 343, history of development, 45960 complexity of functions, 878
347, 351 impetus for development, 459 sexual activity
5a-reductase isoenzymes, 131 mechanisms of tissue-selective effect on testosterone level, 25960
5a-reductase type 1, 24 actions, 4635 sexual arousal
5a-reductase type 2, 24, 36, 340 misuse in sports, 529 and testosterone, 97
5a-reductase type 2 deficiency, non-steroidal SARMs, 460, 4613 and testosterone level, 94, 96
36, 161 potential therapeutic use, 459 sexual desire and response (men)
5a-reductase type 2 inhibitors, 168 preclinical studies with first- and testosterone level, 90
rehabilitation after surgery generation non-steroidal SARMs, components of sexual arousal, 90
androgen therapy, 3846 4656 effects of testosterone, 903
Reifenstein, E.C., 10 prohibition in sports, 529 experimental reduction of
relaxin, 21 regulatory challenges to testosterone, 923
religious reasons for castration, 3 development, 466 increasing testosterone in eugonadal
REM sleep role of co-regulator proteins, 4635 men, 923
nocturnal erections, 2557 sports drug testing, 52930 testosterone replacement in
renal disease steroidal SARMs, 4601 hypogonadism, 902
androgen therapy, 37780 structural classes, 460 testosterone threshold theory, 90
renal failure, 343 schizophrenia, 393, 445, 447 visual erotic stimuli, 91
renal function Scoptic sect, 3 sexual desire and response (women)
effects of androgen therapy, 378 scrotal testosterone patches, 9, 296 activational effects of testosterone,
renal insufficiency, 356 scrotum 98106
renal transplantation, 377 fetal development, 36 central arousal, 98
renin-angiotensin-aldosterone axis, seasonal breeding in mammals, during lactation, 100
220 2712 effects of antiandrogens, 101
reproductive function selective androgen receptor modifiers effects of bilateral ovariectomy,
effects of CAG repeat androgen (SARMs), 287 1023
receptor polymorphisms, 445 self-emulsifying drug delivery system effects of hysterectomy, 1023
respiratory disease (SEDDS) effects of oral contraceptive use,
androgen therapy, 38991 for testosterone undecanoate, 314 1001
retinal atherosclerosis, 210 semen genital response, 98
retinol receptors, 25 Artistotles theory of, 2 incentive motivation, 98
Reynauds phenomenon seminal emission influence of the menstrual cycle, 99
androgen therapy, 260, 383 effects of testosterone withdrawal, 91 masturbation, 99
rhesus monkeys, 89 seminal vesicle, 24 oral contraceptive use, 99
rheumatic diseases, 343 seminiferous tubules, 124, 126 orgasm, 98
rheumatoid arthritis, 439 senescence. See aging ovarian androgens, 100
androgen therapy, 3812 sensitive window patterns of sexuality in women, 106

565
Index

sexual desire and response (women) small intestine muscle non-genomic pathways of androgen
(cont.) effects of androgens, 28 receptor action, 136
problems of low sexual desire, 99 smoking primary spermatocytes, 124
role of estradiol, 98106 and cardiovascular disease, 207 pubertal initiation in rodents, 131
role of testosterone, 98106 and testosterone levels in elderly role of androgens, 1308
sexuality of older women, 100 men, 342 role of androgens in initiation, 128
testosterone deficiency in women, somatic mosaicism role of androgens in maintenance,
98 androgen receptor gene mutations, 41 128
vaginal pulse amplitude (VPA), 98 somatotropic axis, 345 role of the Sertoli cells, 124, 126,
sexual differentiation of embryo and sonic hedgehog (Shh), 37, 252 1325
fetus, 357 Sox9 expression, 345 sites of androgen action, 1325
Sexual Experience Scale, 92 Spallanzani, Lazzaro, 2, 5 species-specific features, 126
sexual interest specificity-affecting androgen receptor spermatid development, 134
influence on erectile function, knock-in mice, 135 spermatids, 124, 134
2545 sperm spermatogonia, 124, 140
sexual offenders first descriptions of, 5 spermatogonial development, 1345
contraindication for testosterone sperm concentrations spermiation, 124
therapy, 303 influence of CAG repeat AR spermiation failure, 134, 138
sexual orientation, 4289 polymorphisms, 45 spermiogenesis, 124, 1378
sexuality normal range, 45 suppression by anabolic steroids,
influence of estrogens in men, sperm cryopreservation 540
4289 Spallanzani, Lazzaro, 5 suppression by testosterone therapy,
monitoring testosterone therapy, sperm maturation, 21 304
298 spermatic vein, 23 testicular androgen production and
sexuality in women spermatids, 124 metabolism, 1302
relationship to mood, 107 development, 134 testicular androgen receptor, 1325
sexually dimorphic development, 357 release from Sertoli cells, 134 treatment implications for male
SHBG (sex hormone-binding spermatogenesis, 1234 hormonal contraception, 144
globulin), 15, 21, 24, 130, 423 adult spermatogenesis, 12930 treatment of secondary
and bone loss in men, 178 and FSH, 13840 hypogonadism, 144
and insulin resistance, 2401 and testosterone, 12730 spermatogonia, 124, 140
binding properties, 60 and testosterone levels, 28 spermatozoa
effects of aging, 3389, 345 aromatization of testosterone to progesterone receptors, 23
effects of HRT in women, 104 estradiol, 1434 spermiation, 124
in women, 100 basic and common features, 1246 spermiation failure, 138
levels in blood, 602 cooperative effects of androgen and spermiogenesis, 124, 1378
levels in elderly men, 3412 FSH, 1403 spinobulbar muscular atrophy
levels in women, 4956 effects of aging, 3367 (XSBMA). See Kennedy
SHBG test, 40 effects of progestogens, 92 syndrome
short tandem repeats (STRs), 25 effects of testicular androgen spironolactone, 168, 169, 343, 537
Silber, Sherman J., 7 concentration, 1378 sports doping. See anabolic androgenic
sildenafil, 257, 2589 evaluation of the process, 126 steroids; SARMs
Sjgrens syndrome genomic pathway of androgen SRD5A inhibitors, 2812
androgen therapy, 383 receptor action, 1356 role in prostate cancer prevention,
skin germ cell development, 1245 2856
genital, 24 hypothalamo-hypophyseal- SRD5A isoenzymes, 2734
non-genital, 24 testicular axis, 1267 see also 5a-reductase.
skin healing in primates, 126 SRD5A1 gene, 165
androgen therapy, 3856 in rodents, 126 SRD5A2 gene, 165
slaves influence of CAG repeat AR SRY (sex-determining region of the
castration, 23 polymorphisms, 445 Y chromosome), 34
sleep intratesticular testosterone, 128, Stanley, Leo, 7
effects of testosterone in men, 967 1302 stanozolol, 40, 518, 520, 524, 538
sleep apnea, 3001, 342, 356, 375 mechanisms of androgen action in StAR expression, 21
and testosterone level, 96 the testis, 1356 StAR gene, 19
androgen therapy, 3901 meiotic arrest, 137 State Self-Esteem Scale, 96
contraindication for testosterone meiotic divisions, 124 statin drugs, 216
therapy, 417 mitotic divisions, 124 statin therapy, 241
sleep-related erections (SREs), 2557 neonatal androgen secretion, 1278 Steinach, Eugen, 7, 8

566
Index

stem cell factor (SCF), 163, 164 testicular feminized mice, 212, 215, testosterone in the blood
stem cells, 254 217, 240 free and protein-bound forms, 602
properties of, 279 testicular intersitium testosterone isocaproate, 319
stem Leydig cells, 16 Leydig cells, 15 testosterone levels
stereology, 126 testicular lymphatic circulation, 22 and cardiovascular risk, 2089
steroid biochemistry, 8 testicular morphology predictive marker for mortality,
steroid-induced osteoporosis, 384 first descriptions of, 5 2246
steroid receptors testicular organotherapy, 67 testosterone microcapsules, 323
evolution of, 271
steroid sulfatase (STS), 439, 440 testis, 24

Testifortan , 6 testosterone patches, 9, 62, 3246
non-scrotal, 296, 325
steroidogenesis, 15 testis cords, 35 scrotal, 296, 3245
adrenal glands, 16 testis development, 345 testosterone pellet implants, 296,
Leydig cells, 1516 testis sicca, 6 3234
steroidogenic acute regulatory (StAR) testis transplantation, 78 testosterone pellets, 474
protein, 1819 testosterone testosterone phenylpropionate, 319
steroids 5a-reduction, 23, 24 testosterone preparations
androgens, 15 aromatization to estradiol, 3501 buccal administration, 31415
discovery of ring structure, 8 chemical synthesis in the twentieth derived from androstane, 31011
stroke, 217, 342 century, 810 fluoxymesterone, 313
structural brain imaging studies, 88 clinically available forms, 910 goal of testosterone therapy, 30910
stump-tailed macaque evolution of reproductive regulatory hepatotoxic side-effects, 312
androgenetic alopecia, 163, 164 role, 2702 history of clinical use, 30912
subdermal testosterone pellet first-pass metabolism in the liver, in castor oil, 3223
implants, 3234 310, 314 in tea seed oil, 3212
sublingual administration of isolation of, 9 intramuscular administration,
testosterone, 31415 levels in depressed women, 107 31523
sudden cardiac death normal range in men, 90 mesterolone, 313
and anabolic steroids, 208 testosterone and gender differences, 17a-methyltestosterone, 312, 314
sudden death caused by thrombosis, 11012 nasal administration, 315
213 testosterone binding protein (TeBG), oral administration, 31013
supraoptic nuclei, 90 273 pharmacokinetic studies, 9
surgery rehabilitation testosterone biosynthesis, 1619 pharmacokinetics of testosterone
androgen therapy, 3846 conversion of cholesterol, 1617 esters, 31516
surgical trauma D5 pathway, 1617 range of approaches, 310
effects on testosterone levels, 342 D4 pathway, 1617 rectal administration, 315
Sustanon, 97 regulation by factors other than subdermal testosterone pellet
Sveinsson, Jonas, 7 LH, 21 implants, 3234
Swammerdam, Jan, 5 regulation by LH, 1920 sublingual administration, 31415
Swedish guidelines on testosterone testosterone buciclate, 3201, 473 synthetic androgens, 310
deficiency, 413 testosterone cream, 303 testosterone buciclate, 3201
Symptom Checklist-90-R, 95 testosterone cyclohexanecarboxylate, testosterone
synthetic androgens, 310 31819 cyclohexanecarboxylate, 31819
systemic lupus erythematosus (SLE), testosterone cypionate, 11, 95, 217, testosterone cypionate, 31819
439, 4456 295, 303, 31819 testosterone decanoate, 323
androgen therapy, 3823 testosterone decanoate, 319, 323 testosterone enanthate, 31718
systemic sclerosis testosterone deficiency symptoms, 294 testosterone ester combinations,
androgen therapy, 383 testosterone enanthate, 9, 92, 94, 96, 31920
193, 211, 295, 303, 31718, testosterone esters, 315
tadalafil, 257, 259 31920, 4723 testosterone gel, 3258
T-cells testosterone esters, 295 testosterone microcapsules, 323
effects of testosterone, 28 forms misused in sports, 520 testosterone patches, 3246
tea seed oil in testosterone pharmacokinetics, 31516 testosterone propionate, 31617,
preparations, 3212 testosterone gel, 10, 46, 62, 195, 295, 31920
temporal cortex, 89, 90 297, 3258 testosterone topical solution, 330
testicular androgen production and advantages over patches, 327 testosterone undecanoate, 31314,
metabolism, 1302 risk of interpersonal transfer, 327, 3213
testicular androgen receptor, 1325 328 transdermal administration,
testicular estradiol, 132 scrotal application, 329 32430
testicular feminization, 10, 27 testosterone immunoassays, 9 unmodified testosterone, 31012

567
Index

testosterone propionate, 9, 303, 315, symptom-specific thresholds for behavior, 297302


31617, 31920 treatment, 297 blood pressure, 299
testosterone rebound therapy, 303 to reduce final height, 3023 bone mass, 301
testosterone replacement transdermal route, 62 cardiac function, 299
effects of testosterone withdrawal, use in types of hypogonadism, erythropoiesis, 3001
902 2924 see also androgen therapy. gonadotropins, 300
effects on muscle mass, 1923 testosterone therapy gynecomastia, 2989
hypogonadal men, 902 contraindications, 3034 lean body mass, 298
timing of effects, 98 breast cancer, 303 lipid metabolism, 301
testosterone therapy patients who wish to father liver function, 301
adverse events in older men, 1956 children, 304 male sexual hair pattern, 298
age-related differences in response, prostate carcinoma, 303 monitoring for prostate carcinoma,
193 reduced spermatogenic function, 301
and prostate cancer, 10 304 mood, 297302
aromatase deficiency, 24 sexual offenders, 303 penis growth, 299
cardiac effects, 221 testosterone therapy for women, phenotype, 2989
choice of preparation, 2947 5005 prostate-specific antigen (PSA), 301
delayed puberty in boys, 302 adverse androgenic effects, 505 prostate volume, 301
effects in healthy older men, 1946 amenorrheic women, 104 sebum production, 298
effects on chronically ill patients, assessing women with low sexual serum DHT, 300
1934 desire, 5089 serum estradiol, 300
effects on depression, 1967 breast cancer risk, 5056 serum testosterone, 299300
effects on fatigue/energy, 1967 effects on bone mineral density, 504 sexuality, 298
effects on mood, 1967 effects on cardiovascular function, sleep apnea, 3001
effects on quality of life, 1967 504 voice mutation, 299
effects on well-being, 1967 effects on cardiovascular risk, 504 testosterone-to-epitestosterone ratio,
for micropenis, 303 effects on cognition, 5045 5289
for microphallus, 303 effects on insulin resistance, 504 drug testing in sport, 519
goal of, 30910 effects on lean mass, 504 testosterone-to-estradiol ratio, 427
guidelines for monitoring patients, effects on mood, 504 testosterone topical solution, 330
417 effects on well-being, 504 application site erythema, 330
guidelines on potential risks, endometrial cancer risk, 5067 risk of interpersonal transfer, 330
41617 female sexual dysfunction (FSD), testosterone transport, 213
HIV patients, 1934 5004 entry into target cells, 22
ineffective applications, 303 following bilateral ovariectomy, free testosterone in serum, 21
influence of CAG repeat AR 1056 testosterone undecanoate, 9, 91, 96,
polymorphisms, 478 healthy pre-menopausal women, 104, 218, 242, 244, 295, 296, 303,
level for initiation of therapy, 297 103 31314, 315, 3213
long-term effects, 348 hypoactive sexual desire disorder in castor oil, 4734
mechanisms of effects on muscle, (HSDD), 5004 in tea seed oil, 473
1979 issues related to, 4945 intramuscular form, 10
overall effect, 304 naturally postmenopausal women, oral, 473
potential benefits for hypogonadal 1045 self-emulsifying drug delivery
men, 304 need for an approved formulation system (SEDDS), 314
potential use to improve physical
function, 200
for women, 509
premenopausal women with

Testoviron Depot, 319, 320
tetrahydrogestrinone (THG), 522
range of potential benefits, 2924 endocrine abnormalities, 104 tetrahydroquinoline analogs, 461
role of growth hormone, 199 premenopausal women with sexual tetrahydroquinoline derivatives, 462
role of IGF-1, 199 problems, 1034 TGF-b (transforming growth factor
routes of administration, 2957 safety concerns, 5057 beta), 164
side effects, 24 surgical menopause, 1056 TGG repeats (polyproline), 25
strategies for selective action, transdermal testosterone, 503 threshold level of testosterone, 98
197200 treatment options, 509 thrombocytopenia
strategies to avoid adverse effects, vulval lichen sclerosis treatment, androgen therapy, 377
197200 505 thrombosis formation, 21516
suppression of spermatogenesis, women likely to benefit, 5078 thrombotic occlusion of arteries, 217
304 women using HRT, 1045 thrombus formation, 213
symptoms of testosterone testosterone therapy surveillance, thyroid hormones, 21, 341
deficiency, 294 297302 effects in elderly men, 342

568
Index

thyroid hormone receptors, 22, uterus, 24 causes of low testosterone, 4968


25, 62 development of, 35 cognitive function and testosterone,
thyroid-stimulating hormone, 341 500
thyrotoxicosis, 62 vagina effects of adrenalectomy, 496
tibolone, 541 development of, 35 effects of hormone replacement
tight junctions vardenafil, 257 therapy (HRT), 496
between Sertoli cells, 135 varicocele, 292 effects of hyperprolactinemia, 498
tissue plasminogen activator (tPA), Vasari, Giorgio, 2 effects of hypopituitarism, 498
215 vascular disease effects of hypothalamic
TNF-a (tumor necrosis factor alpha), androgen therapy, 3934 amenorrhea, 498
213, 239, 241, 2445 vascular endothelial growth factor effects of hysterectomy, 496
transactivation function (VEGF), 281 effects of oophorectomy, 496
and short tandem repeats, 25 vascular reactivity effects of oral contraceptives,
transcriptional coactivator or co- and low testosterone, 211 496
repressor proteins, 275 influence of testosterone, 21415 effects of premature ovarian failure,
transdermal testosterone vascular tone, 214 498
administration in women, 104 vasoactive intestinal peptide (VIP), endogenous testosterone and health,
transdermal testosterone gel. 255 498500
See testosterone gel Vatican osteoporosis and testosterone,
transdermal testosterone patches, 9, castrato singers, 4 499500
223, 295 venous thromboembolism risk, postmenopausal ovarian
transdermal testosterone preparations, 21516 testosterone, 496
2967, 32430 ventromedial hypothalamic nuclei, 89 sexual function and testosterone,
transgender identity, 88 vertebral fractures in elderly men, 350 498
transgendered individuals veterans SHBG levels, 4956
effects on mood and aggression, 109 cardiovascular risk, 255 testosterone physiology in
transplantation violence in women, 1089 reproductive years, 4956
testicular, 78 voice mutation, 299 testosterone therapy issues,
transsexuals, 88 Voronoff, Serge, 7 4945
trauma Voss, H.E., 8 well-being and testosterone, 499
androgen therapy, 3846 vulval lichen sclerosis see also testosterone therapy for
trenbolone, 521 testosterone therapy, 505 women.
trenbolone acetate, 521 World Anti-Doping Agency (WADA),
triglycerides, 244, 347 Wagner, Rudolph, 6 519, 520, 5234
role in atherogenesis, 217 wasting
tuberculosis, 193 androgen therapy, 394 X chromosome
tumors of gonads weight loss androgen receptor gene, 25
in AIS, 39 effects of testosterone therapy, 221 XSBMA. See Kennedy syndrome
Turner syndrome, 504 well-being
effect of DHEA treatment, 4424 Y chromosome
ubiquitination of histones, 26 effects of testosterone therapy, SRY (sex-detemining region of the
ulcerative colitis, 446 1967 Y chromosome), 34
ultradian pattern of episodic effects of testosterone therapy for yohimbine, 6, 261
testosterone secretion, 340 women, 504 Young Mania Rating Scale (YMRS),
ultrafiltration method, 76 William the Conqueror, 3 95
unilateral anorchia, 2 Wolffian ducts, 35
unmodified testosterone, 31012 women zoledronic acid, 183
urogenital epithelium, 36 cardiovascular disease and zona reticularis, 438
urogenital sinus, 356, 272, 2778 testosterone, 499

569
(A) (B)
LDL
Steroid carrier
LDL receptor

Steroid
receptor
Steroid
Endosome

Nucleus
Cholesterol

Lysosome
Intracellular Intracellular

Extracellular

(C) Steroid carrier (D)


Steroid carrier

Megalin Steroid channel

Steroid
receptor

Endosome Nucleus Steroid

Nucleus
Intracellular Intracellular

Fig. 2.5 Mechanisms by which steroidal hormones can enter cells. (A) The steroid hormone diffuses freely across the cell membrane, binds to
an intracellular receptor and enters the nucleus to regulate gene expression. (B) Receptor-mediated endocytosis of steroid containing lipophilic
molecules. The LDL binds its receptor and is taken up, degraded in lysosomes and the steroid cholesterol can enter different metabolic
pathways. (C) Receptor-mediated endocytosis of steroids. The entire hormone carrier is endocytotically bound after binding to a carrier protein.
Following the intracellular degradation of the carrier, the ligand hormone is released into the free cytoplasm. (D) Transport-mediated uptake of
molecules through the membrane. The steroid carrier is recognized by a membrane receptor and the ligand is transported into the cell.
(A) Hair changes during the human life cycle

Baby Childhood Puberty Reproductive Stages Aging Death

(B) Abnormal hair growth

Androgen insufficiency
syndromes

XY XY Female androgenetic
no androgen 5 -reductase Hirsutism alopecia
receptors deficiency Gradual thinning on
Child hair Female hair Male hair vertex retention of
pattern only distribution distribution frontal hairline
Fig. 7.1 Human hair varies with life stage and endocrine state. (A) Changes in hair distribution and color signal a persons age, state
of maturity and sex. Visible (i.e. terminal) hair with protective functions normally develops in children on the scalp, eyelashes and eyebrows.
Once puberty occurs, more terminal hair develops on the axilla and pubis in both sexes, and on the face, chest, limbs and often back in
men. Androgens also stimulate hair loss from the scalp in men with the appropriate genes in a patterned manner, causing androgenetic
alopecia. As age increases hair follicles lose their ability to make pigment, causing graying (canities). (B) People with various androgen
insufficiency syndromes demonstrate that none of this occurs without functional androgen receptors and that only axillary and female
pattern of lower pubic triangle hairs are formed in the absence of 5a-reductase type 2. Male pattern hair growth (hirsutism) occurs in women
with abnormalities of plasma androgens or from idiopathic causes, and women may also develop a different form of hair loss, female
androgenetic alopecia or female pattern hair loss (FPHL).
Long, thick Fig. 7.2 Androgens have paradoxically different effects on
(A) BEARD pigmented hair human hair follicles depending on their body site. (A) During
Short, fine
unpigmented
and after puberty, androgens stimulate the gradual
hair transformation of small follicles producing tiny, virtually
+ colorless, vellus hairs, to terminal follicles producing longer,
ANDROGENS
thicker and more pigmented hairs. These changes involve
passing through the hair cycle (see Fig. 7.3). (B) At the same
time, many follicles on the scalp and eyelashes continue to
produce the same type of hairs, apparently unaffected by
Vellus Terminal androgens. (C) In complete contrast, androgens may inhibit
follicle follicle follicles on specific areas of the scalp in genetically
susceptible individuals, causing the reverse transformation of
(B) NON-BALDING SCALP terminal follicles to vellus ones, and androgenetic alopecia.
Androgen
independent (Diagram reproduced from Randall 2000a.)
Long, thick
pigmented hair

+
ANDROGENS

blood
vessels

(C) BALDING SCALP


Androgen
sensitive
Long, thick
pigmented hair
Short, fine
unpigmented hair

+
ANDROGENS

Terminal Vellus
follicle follicle

BALDING SCALP: androgen sensitive

+ ANDROGENS + ANDROGENS

LONG, THICK
PIGMENTED
HAIR
New New
hair hair

Club Club
hair Sebaceous hair Sebaceous
gland gland
Hair Hair
germ Original germ Original
New New
hair Dermal hair
hair hair
Dermal papilla
papilla Dermal cells
cells papilla
Dermal
papilla
Dermal
papilla
TERMINAL
FOLLICLE

Fig. 7.3 Androgen inhibition of hair size on the scalp leads to balding. Hair follicles pass through regular cycles of growth (anagen),
regression (catagen), rest (telogen) and hair shedding during which the lower part of the follicle is regenerated. This enables the follicle to
produce a different type of hair in response to hormonal stimuli to coordinate to changes in the bodys development, e.g. sexual maturity or
seasonal climate changes. Androgens inhibit follicles on specific areas of the scalp in genetically susceptible individuals, causing the
regenerated follicles to be smaller, protrude less into the dermis and produce smaller, less-pigmented hairs. Eventually the terminal hairs of
childhood and early adulthood are replaced by the vellus hairs of androgenetic alopecia, and the area appears bald. Follicles must pass
through a full hair cycle, probably a succession of cycles, to accomplish major changes. (Diagram reproduced from Randall 2010.)
Sex hormone
Hair

Hair bulb
Basement membrane Melanocyte Osteoblast
(indirect target)
?
? Extracellular matrix
RANKL
Epithelial cells ? +T OPG
(indirect target) +T ?
+T Dermal papilla cells
(direct target)
RANK
?
?
Blood capillary T Osteoclast c-fms M-CSF
T
precursor
T
T CIRCULATING ANDROGENS T T

Fig. 7.5 Current model of androgen action in the hair follicle: in the
current, well-accepted hypothesis, androgens from the blood enter
the hair follicle via the dermal papillas blood supply. If appropriate, Mature
they are metabolized to 5a-dihydrotestosterone. They bind to osteoclast
androgen receptors in the key regulatory cells of the follicle, the
dermal papilla cells causing changes in their production of
regulatory paracrine factors; these then alter the activity of dermal BONE
papilla cells, follicular keratinocytes and melanocytes.
T = testosterone; ? = unknown paracrine factors. (Modified from Fig. 8.1 A schematic overview of the RANKL/RANK/OPG system.
Randall 2000b.) Receptor activator of nuclear factor-kB ligand mediates a signal for
osteoclast formation through RANK expressed on osteoclast
progenitors. Osteoprotegerin counteracts this effect by competing
for and neutralizing RANKL. Osteoclast differentiation factor is a
ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor.
M-CSF is an essential factor for osteoclast proliferation and
differentiation, which is produced by osteoblasts in osseous tissues
and acts through its receptor c-fms.

Multipotent
progenitor cells

Satellite cells

Differentiated
myocyte

OH
AR im IGF-1
CH CH3
CH3
CH3

SRD5A O
O H IGF-1R
DHT
T Muscle
protein
Atrogens/FoXo synthesis
Ubiquitin/
proteasome

GH IGF-1
Fig. 9.3 Potential mechanisms of the anabolic effects of testosterone on the skeletal muscle. Testosterone (T) could potentially increase
muscle mass through multiple mechanisms. The vast majority of evidence suggests that testosterones effects are mediated through the AR.
It does not appear that 5-alpha reduction of testosterone is essential for mediating testosterones effects on the muscle. Testosterone stimulates
GH secretion and increases IGF-1 levels, but circulating GH and IGF-1 are not essential for mediating testosterones effects on the muscle.
Testosterone promotes the differentiation of mesenchymal multipotent progenitor cell into the myogenic lineage. Whether testosterone has
direct effects on muscle protein synthesis, muscle protein degradation or atrophy pathways remains to be investigated. Testosterones effect on
proliferation and differentiation of skeletal muscle progenitor cells requires the mediation of IGF-1R signaling.
Belarus
Ukraine
Kazakstan
Azerbaijan
Russian Federation
Krygyzstan
Lithuania
Hungary
Romania
Czech Republic
lreland
Finland
UK
New Zealand
Austria
USA
Norway
Sweden
Denmark
Australia
Greece
Netherlands
Luxembourg Men
Italy
Spain Women
France
Japan

Fig. 10.1 Figure showing the consistent sex difference in death rates for coronary artery disease across both high- and low-prevalence
communities. Data are for men and women aged 3574 for 1998, from the BHF coronary statistics database released in 2002.

IDF NCEP ATP III Fig. 11.1 Definitions of the metabolic syndrome which are
in common usage. *These two definitions have recently been
amalgamated, with agreement that three out of the five
Essential: Three factors from:
criteria are required for the diagnosis. As there are ethnic and
*Central obesity waist >94 cm population differences in the degree of elevated waist
*Central obesity waist >102 cm circumference, it is important that this is taken into account
for country-specific measurements. In addition, drug
Plus two from: Fasting glucose >5.6 mmol/l
treatment of elevated triglycerides, blood pressure and
Blood pressure >130/85 fasting glucose and reduced HDL-C, even if normalized for
Fasting glucose >5.6 mmol/l Fasting triglycerides >1.7 mmol/l each criterion, are included (Alberti et al. 2009).
Blood pressure >130/85 HDL cholesterol <1.03 mmol/l
Fasting triglycerides >1.7 mmol/l
HDL cholesterol <1.03 mmol/l

(4)

Estradiol

Hypothalamic-
TNF-
pituitary axis
IL-6

+
Leptin
(3)

Insulin
LH pulse
resistance
+ amplitude

(1)
Adipocyte number Testis
and size Aromatase

Adipose
Triglyceride tissue Testosterone
uptake

Lipoprotein lipase
activity

(2)

Fig. 11.2 The Hypogonadal-Obesity-Adipocytokine Hypothesis. (1) High aromatase activity in adipocytes converts testosterone to estradiol.
Reduced tissue testosterone facilitates triglyceride storage in adipocytes by allowing (2) increased lipoprotein lipase activity and stimulating
pluripotent stem cells to mature into adipocytes. (3) Increased adipocyte mass is associated with greater insulin resistance. (4) Estradiol
and adipocytokines TNF-a, IL-6 and leptin (as a result of leptin resistance in human obesity) inhibit the hypothalamic-pituitary-testicular axis
response to decreasing androgen levels (blue arrows). Orange arrow, hypogonadal obesity cycle; green arrow, low testosterone promotes
the formation of adipocytes from pluripotent stem cells; +, positive effect; , negative effect. (From Jones 2010).
(B) Testosterone
OH
CH3
(A) Sympathetic nerve NANC nerves CH3

Ca2+ eNOS Endothelial cell


O
NA CH3 OH nNOS
CH3 NO Ca2+
NO
VOC CLCAs 1 O
RhoA-GTP Testosterone
Smooth (active) Smooth Hyperpolarization Ca2+-ATPase
muscle cell GEFs GAPs muscle cell NO K+ pump
PLC RhoA-GDP
(inactive) sGC InsP3
ROCK cGMP PKG
ATP
InsP3
ATP ATP GTP
CPI-17 Ca2+ Ca2+
Ca2+ Ca2+ GMP PDE5 Ca2+ 2+
Ca
Ca2+ 2+
Ca P Ca2+
Ca2+ MLCP
MLC MLC SR
SR CH3 OH P
MLCK CH3 MLCK
Ca2+/calmodulin complex Activation MLC MLC
Activation MLCP
Inhibition O
Inhibition
CONTRACTION Testosterone
RELAXATION

Fig. 12.1 Schematic representation of the biochemical events leading to penile flaccidity (A) or erection (B). The enzymatic steps for which a role for testosterone has been proposed are
represented. (A) Noradrenalin (NA) binding generates inositol 1,4,5-trisphosphate (InsP3), which, by increasing intracellular calcium (Ca2+) levels, activates Ca2+-sensitive chloride channels (CLCAs),
resulting in membrane depolarization with the diffusion of the stimulus to the neighboring cells and the opening of voltage-operated channels (VOC). The increased Ca2+ flow promotes,
through calmodulin, activation of myosin light chain (MLC) kinase (MLCK) and cell contraction. Cell contraction is also obtained by altering the Ca2+ sensitivity through a NA-induced activation of a
second pathway, RhoA/ROCK, which increases, through a series of kinase activation, the sensitivity of MLC to Ca2+. Testosterone is supposed to negatively regulate the latter event. (B) Nitric oxide
(NO) is generated by NO synthases in either non-adrenergic, non-cholinergic (NANC) neurons (nNOS) or endothelial cells (eNOS). Both steps are positively regulated by testosterone. Nitric oxide
diffuses into SMC and activates a soluble guanylate cyclase (sGC), which in turn transforms GTP into cGMP. Cyclic GMP activates protein kinase G (PKG), which, through the indicated pathways, finally
decreases intracellular Ca2+ levels, leading to relaxation. Phosphodiesterase type 5 (PDE5) metabolizes cGMP into GMP, thereby limiting its effects. The latter event is positively controlled by
testosterone. Other abbreviations: cGMP, cyclic GMP; CPI-17, protein phosphatase 1 regulatory, subunit 14A; GAP, GTPase activating protein; GEF, guanine nucleotide exchange factor; MLCP, myosin
light chain phosphatase; PLC, phospholipase C; rhoA, ras homolog gene family member A; ROCK, rho-associated, coiled-coil containing protein kinase; SR, sarcoplasmic reticulum.
Fig. 14.1 Threshold levels of serum
testosterone for various symptoms of
Total testosterone (nmol / l) late-onset hypogonadism (LOH) in
434 patients (Zitzmann et al. 2006).
20

74
Normal

69
15

Loss of libido
84
Loss of vigor
12
Obesity 65
10
Feeling depressed, disturbed sleep
Lacking concentration 67
Diabetes mellitus type 2 (also non-obese men)
8
Hot flushes
Erectile dysfunction 75

0
(A) Ligand-bound (C) Residue mobility difference 1.0 A

1.0 A

(B) Apo-form (D) Residue group dynamics

15
Non-CoaBC

CoaBC
10
RMSD (A)

CoaRS2

5 CoaRS1

LBI
0
0 2 4 6 8 10
Time (ns)

Fig. 21.3 Molecular modeling of AR-LBD upon ligand binding. The simulations were performed at 295.5 K and atmospheric pressure.
(A, B) Superimposed representations of AR-LBD in the ligand-bound form and apo-form, respectively. Pink coloring shows the initial structures,
and the pale green coloring shows the structures after 10 ns of simulations. (C) The AR-LBD apo-form colored by the difference between
standard deviations of residue RMSDs (root mean square deviations) for LBD-R1881 complex and apo-form over 610 ns. Positive values
correspond to greater mobility in the apo-form. (D) The predicted RMSDs for ligand-binding interface (LBI), coactivator recruitment surface
1 (CoaRS1), coactivator recruitment surface 2 (CoaRS2) and coactivator binding cleft (CoaBC). Red curves show RMSDs of helices in a ligand-
bound structure, green curves in an apo-form structure. The groups of residues are defined as follows: LBI (V685, L701, N705, L707, Q711,
L744, M745, M749, R752, Y763, F764, Q783, M787, F876, T877, L880, F891, M895), CoaRS1 (V713, V730, M734), CoaRS2 (E709, L712, V713, V715,
V716, K717, K720, F725, R726, V730, Q733, M734, I737, Q738), CoaBC (helices 3, 30 , 4, 5, 12), Non-CoaBC (helices 1, 6, 7, 8, 9, 10, 11). Reproduced
with permission from Jasuja et al. (2009).

Percentage of Fig. 23.4 Effects of different doses of the


each treatment transdermal testosterone patch on androgenic
group affected events over 12 months in postmenopausal
25 women not using concurrent estrogen therapy.
Placebo Adapted from Davis et al. (2008a).

20 150 g TTP
300 g TTP
15

10

0
Acne Increased hair Alopecia Voice change
growth

Anda mungkin juga menyukai