Anda di halaman 1dari 12

The new england journal of medicine

review article

medical progress

b-Thalassemia
Deborah Rund, M.D., and Eliezer Rachmilewitz, M.D.

t halassemia is a hereditary anemia resulting from defects in


hemoglobin production.1 b-Thalassemia, which is caused by a decrease in the
production of b-globin chains (Fig. 1), affects multiple organs and is associ-
ated with considerable morbidity and mortality.2 Accordingly, lifelong care is required,3
and financial expenditures for proper treatment are substantial.4
From the Hematology Department, He-
brew University–Hadassah Medical Cen-
ter, Ein Kerem, Jerusalem (D.R.); and the
Hematology Department, E. Wolfson
Medical Center, Holon (E.R.) — both in
Israel. Address reprint requests to Dr.
Rund at the Hematology Department, Ha-
Thalassemia is among the most common genetic disorders worldwide; 4.83 per- dassah University Hospital, Ein Kerem,
cent of the world’s population carry globin variants, including 1.67 percent of the pop- Jerusalem IL91120, Israel, or at rund@
ulation who are heterozygous for a-thalassemia and b-thalassemia. In addition, 1.92 cc.huji.ac.il.

percent carry sickle hemoglobin, 0.95 percent carry hemoglobin E, and 0.29 percent N Engl J Med 2005;353:1135-46.
carry hemoglobin C. Thus, the worldwide birth rate of people who are homozygous or Copyright © 2005 Massachusetts Medical Society.
compound heterozygous for symptomatic globin disorders, including a-thalassemia
and b-thalassemia, is no less than 2.4 per 1000 births, of which 1.96 have sickle cell
disease and 0.44 have thalassemias.5

molecular and cellular pathology

b-Thalassemia is caused by any of more than 200 point mutations and, rarely, by dele-
tions.1 Thalassemia is clinically heterogeneous because various genetic lesions vari-
ably impair globin-chain synthesis. However, genotypic variability at known loci is of-
ten insufficient to explain the disparate phenotypes of individual patients with the
same genotype. Disparity between genotypes and phenotypes is particularly marked in
thalassemia intermedia and hemoglobin E thalassemia (Table 1). However, the known
genetic factors are insufficient to account for the marked variability, and other genetic
modifiers may exist.6
Recently, an a-hemoglobin–stabilizing protein was identified that binds to and sta-
bilizes free a chains, thereby blocking the production of reactive oxygen species and
reducing oxidative damage to erythrocytes. This protein appears to modulate the clini-
cal picture of b-thalassemia in a murine model7 but in human studies has not been
found to modify thalassemia.
Hemolysis and ineffective erythropoiesis together cause the anemia that occurs in
thalassemia. The relative contributions of these two pathologic processes differ in var-
ious forms of thalassemia.8 Figure 2 illustrates the complex chain of events that occurs
in erythrocytes, resulting in their accelerated peripheral destruction.
The bone marrow of patients with thalassemia contains five to six times the number
of erythroid precursors as does the bone marrow of healthy controls,17 with 15 times
the number of apoptotic cells in the polychromatophilic and orthochromic stages.17,18
Accelerated apoptosis, the major cause of ineffective erythropoiesis, is caused by ex-
cess a-chain deposition in erythroid precursors.8 Although the exact mechanism is not
known, a death-receptor–mediated pathway seems to be involved with Fas–Fas ligand
interactions.19 In normal erythropoiesis, apoptotic mechanisms seem to play a regula-
tory role and are required for normal erythroid maturation.20 Accelerated apoptosis is

n engl j med 353;11 www.nejm.org september 15, 2005 1135

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Figure 1. Management of Thalassemia and Treatment-Related Complications.


The anemia that is associated with thalassemia may be severe and is accompanied by ineffective erythropoiesis, with bone
expansion and extramedullary hematopoiesis in the liver, spleen, and other sites, such as paravertebral masses. Transfu-
sion therapy, which is the mainstay of treatment, allows for normal growth and development and suppresses ineffective
erythropoiesis. Transfusion-transmitted infections (primarily hepatitis B and C) are an important cause of death in coun-
tries where proper testing is not available. Iron overload results both from transfusional hemosiderosis and excess gas-
trointestinal iron absorption. Iron deposition in the heart, liver, and multiple endocrine glands results in severe damage
to these organs, with variable endocrine organ failure. The endocrinopathies can be treated with hormone replacement.
However, the most serious result of iron overload is life-threatening cardiotoxicity, for which chelation therapy is required.
Thalassemia can be cured by bone marrow transplantation. Experimental therapies to ameliorate the anemia that have
been or are currently under investigation include fetal hemoglobin modifiers and antioxidants. In the future, gene thera-
py or other molecular methods may be feasible.

1136 n engl j med 353;11 www.nejm.org september 15 , 2005

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

Table 1. Genetic Basis and Clinical Manifestations of Common b-Thalassemia Syndromes.

Feature Thalassemia Trait Thalassemia Intermedia Thalassemia Major Hemoglobin E Thalassemia


Genetic pathology One b-globin gene carry- Two b-globin genes carrying Two b-globin genes carrying One b-globin gene carrying
ing a thalassemia mu- a thalassemia mutation, a severe thalassemia mu- a thalassemia mutation
tation at least one of which is tation (mild or severe) in com-
mild; one b-globin thal- bination with a b-globin
assemia mutation in gene carrying the point
combination with excess mutation encoding he-
a-globin genes (less moglobin E
common)
Clinical manifesta- Mild or no anemia, with Mild to moderate anemia; Severe anemia requiring reg- Mild to severe anemia;
tions variable microcytosis relative independence ular transfusions begin- relative dependence
(mean corpuscular from transfusions; prom- ning in infancy; spleno- on transfusions; spleno-
volume, 60 to normal); inent splenomegaly and megaly and bone disease megaly and bony defor-
no splenomegaly; no bone deformities; varia- depending on the effica- mities; variable degree
bone disease ble degrees of iron over- cy of transfusion therapy; of iron overload, de-
load depending on sever- severe iron overload pending on severity of
ity of anemia and trans- anemia and transfusion
fusion requirement requirement
Severity Asymptomatic From asymptomatic to se- Lifelong supportive care re- From asymptomatic to se-
verely symptomatic quired verely symptomatic
Ameliorating genet- Presence of concurrent Presence of concurrent Presence of concurrent Mild b-thalassemia muta-
ic factors a-thalassemia a-thalassemia; elevated a-thalassemia; elevated tion; concurrent a-thal-
hemoglobin F hemoglobin F assemia; elevated he-
moglobin F
Exacerbating genet- Excess a-globin genes Number of excess a-globin None known Severe b-thalassemia
ic factors genes (five or more) mutation

associated with a rise in extracellular exposure of ciency.24 Growth hormone therapy has had variable
phosphatidylserine (Fig. 2), an important signal for success.23 Hypogonadotropic hypogonadism im-
removal by activated macrophages,14 whose num- pairs fertility but can be corrected with the use of
bers are increased in thalassemic bone marrow.21 hormonal replacement in male patients. A small
number of female patients, including those with
thalassemia major or thalassemia intermedia, have
clinical manifestations
an d supportive therapy been able to become pregnant, either spontane-
ously (if they have received adequate chelation ther-
anemia and transfusion therapy apy) or with assisted reproductive techniques.23
Regular transfusion therapy to maintain hemo- Pregnancy generally appears safe if baseline car-
globin levels of at least 9 to 10 g per deciliter allows diac function is good.25
for improved growth and development and also Considerable morbidity in older patients re-
reduces hepatosplenomegaly due to extramedullary sults from bone disease due to osteopenia and os-
hematopoiesis as well as bone deformities.2,3 Ta- teoporosis, which is often accompanied by disabling
ble 2 summarizes some of the advances in transfu- pain and fractures. The pathogenesis is complex
sion therapy. and multifactorial. Bone marrow expansion due to
ineffective erythropoiesis, endocrine dysfunction,
endocrinopathies and bone disease and complications of treatment all contribute to
Impairment of growth and endocrinopathies, par- the condition.26 Overly vigorous chelation is asso-
ticularly hypogonadism, are common features of ciated with deferoxamine-induced bone dysplasia,
thalassemia.2,23,24 Since these manifestations (Fig. which can slow growth velocity in children and may
1) result from chronic anemia as well as from iron be only partially reversible.24 Bone-disease manage-
overload, they are much more common in older ment includes the careful monitoring of chelation,
patients or those whose chelation therapy is insuffi- lifestyle adjustments (increased calcium intake and
cient (discussed below).2,23 Hormonal replace- physical activity and refraining from smoking), hor-
ment is indicated for residual endocrine insuffi- monal therapy, and vitamin D therapy. Osteoclast

n engl j med 353;11 www.nejm.org september 15, 2005 1137

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

inhibitors such as bisphosphonates have the po-


tential to reduce bone resorption and may be a valu- Figure 2 (facing page). Pathophysiological Features
of Hemolysis and Hypercoagulability.
able treatment approach, but further study is re-
quired before the routine use of these drugs can be Oxidation of a-, b-, or g-hemoglobin subunits leads to
the formation of hemichromes, whose rate of formation
recommended.26 determines the rate of hemolysis.9 Because a chains dis-
sociate into monomers more readily than do b or
g chains, they form hemichromes at a faster rate,10
iron overload — pathogenesis, which explains why b-thalassemia is clinically much
measurement, and therapy more severe than a-thalassemia. Precipitates of un-
paired b chains form single large inclusions known as
Iron overload causes most of the mortality and mor- Heinz bodies.11 This process occurs at a later stage of
bidity associated with thalassemia. Iron deposi- maturation than does the formation of smaller multiple
tion occurs in visceral organs (mainly in the heart, inclusions because of the precipitation of unpaired a
chains. Hemichromes bind to or modify various compo-
liver, and endocrine glands), causing tissue dam- nents of the mature red-cell membrane, such as protein
age and ultimately organ dysfunction and failure band 3, protein 4.1, ankyrin, and spectrin.12 After precipi-
(Fig. 1). Cardiac events due to iron overload are still tation of hemichromes, heme disintegrates, and toxic
the primary cause of death.27,28 Both transfusional non–transferrin-bound iron species are released.13 The re-
sulting free iron catalyzes the formation of reactive oxygen
iron overload and excess gastrointestinal absorp-
species.9 Iron-dependent oxidation of membrane proteins
tion are contributory. Paradoxically, excess gastro- and formation of red-cell “senescence” antigens such as
intestinal iron absorption persists despite massive phosphatidylserine14 cause thalassemic red cells to be rig-
increases in total body iron load. id and deformed and to aggregate, resulting in premature
Hepcidin is a small peptide that inhibits iron ab- cell removal.15 Phosphatidylserine is also involved in the
activation of the coagulation system.16
sorption in the small bowel. Hepcidin levels nor-
mally increase when iron stores are elevated.29 Hep-
cidin levels were found to be inappropriately low in rately represent iron deposition in other vital organs
patients with thalassemia intermedia and thalas- (such as the heart). Indeed, severe cardiac damage
semia major.30 Furthermore, serum from patients has been observed in a few patients with presum-
with thalassemia inhibited hepcidin messenger ably adequate chelation, and myocardial iron and
RNA expression in the HepG2 cell line, which sug- left ventricular function apparently cannot be pre-
gests the presence of a humoral factor that down- dicted from liver iron concentration, ferritin levels,
regulates hepcidin.31 These observations suggest or both.34 Therefore, noninvasive techniques for
that the administration of hepcidin or agents that the measurement of cardiac iron levels are being
increase hepcidin expression may be therapeuti- developed. Magnetic resonance imaging (MRI) for
cally useful for the inhibition of inappropriate iron the measurement of cardiac iron is technically prob-
absorption. lematic. However, the application of T2 gradient-
Accurate, preferably noninvasive measurement echo sequencing is more sensitive to hemosiderin
of iron stores is crucial for the evaluation and man- deposition and appears to be useful for the mea-
agement of chelation therapy. Serum ferritin is surement of myocardial iron in thalassemia,35 but
most commonly measured as an indicator of iron this approach requires further validation and long-
stores. Ferritin levels below 2500 mg per milliliter term studies to determine its usefulness in assess-
are associated with improved survival, free of car- ing the effectiveness of chelation therapy.
diac disease.32 However, serum ferritin levels are Elevated tissue iron stores are only one com-
highly unreliable, particularly when liver disease ponent of the damaging effects of iron overload.
is present.33 Liver biopsy has often been used but is A highly toxic form of iron, non–transferrin-bound
invasive. Direct noninvasive measurement of he- iron, is formed when the iron-binding capacity of
patic iron stores is possible with the technique of transferrin has been exceeded. Non–transferrin-
magnetic susceptometry (with the use of a super- bound iron is highly toxic because it can catalyze
conducting quantum-interference device) and is the formation of reactive oxygen species through
either equivalent to or more accurate than the mea- the Fenton reaction.9 A fraction of non–transfer-
surement of hepatic iron by liver biopsy.28 However, rin-bound iron, the labile plasma iron, can be mea-
only four centers worldwide currently have this sured directly and may serve as a clinically useful
capacity. test for monitoring iron-chelation therapy.36
It is noteworthy that hepatic iron may not accu- Iron-chelation therapy is largely responsible for

1138 n engl j med 353;11 www.nejm.org september 15 , 2005

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

n engl j med 353;11 www.nejm.org september 15, 2005 1139

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Table 2. Improvements in Supportive Care of b-Thalassemia.*

Treatment Approach Outcome


Transfusion
Definition of the optimal end point of transfusion Less iron overload and alloimmunization
(a hemoglobin level of 9–10 rather than 10–12)
Use of leukodepletion techniques Fewer febrile nonhemolytic transfusion reactions, less
transmission of cytomegalovirus, less alloimmunization22
Testing for viruses (including hepatitis B and C, Substantial reduction in transfusion-transmitted infections
HIV, and HTLV-I)
Implantation of venous-access device More effective therapy and improved patient comfort
and compliance
Chelation
Individualization of doses of deferoxamine Fewer untoward effects (hearing loss, bone dysplasia)
Development of oral chelators and combined Improved compliance and efficacy
chelation therapy
Endocrine support
Administration of hormone replacement (sex, Improvement in growth, development, and sexual maturation;
thyroid, and growth hormones) prevention of osteoporosis
Administration of fertility agents Induction of spermatogenesis; achievement of pregnancy
Administration of osteoclast inhibitors Improvement in osteopenia, osteoporosis, and quality of life

* HIV denotes human immunodeficiency virus, and HTLV-I human T-cell lymphotropic virus type I.

doubling the life expectancy of patients with thal- iprone may be more effective than deferoxamine in
assemia major.28,32,37 Deferoxamine continues to the removal of myocardial iron.34,43,44
be the most common iron-chelating agent in use, An encouraging new approach to chelation
but it has several limitations: the need for paren- therapy is the sequential combined administration
teral administration (which is painful and reduces of deferiprone and deferoxamine. Experimental evi-
compliance), side effects, and cost (which is pro- dence suggests that intracellular iron chelated by
hibitive in underdeveloped countries).28 deferiprone is transferred in the plasma to the
Much effort has been invested in the develop- more powerful chelator, deferoxamine (the so-called
ment of new orally active chelators. Deferiprone, “shuttle hypothesis”).45 As a consequence, more
an orally administered chelator, was initially thought iron is excreted with the use of combined therapy
to be an inadequate chelator that might worsen he- than with the separate administration of each drug.
patic fibrosis. However, cumulative worldwide ex- Furthermore, the compliance of patients was im-
perience indicates that the drug is safe and effec- proved with the use of combined therapy because
tive.38 Long-term administration of deferiprone fewer painful injections of deferoxamine were re-
does not appear to be associated with liver dam- quired.43,46,47
age.39 Adverse effects of deferiprone include ar- A number of new oral iron chelators are under
thralgia, nausea and other gastrointestinal symp- development.25 Deferasirox (ICL670) is particu-
toms, fluctuating liver enzyme levels, leukopenia, larly promising for its efficacy, which may be sim-
and rarely agranulocytosis and zinc deficiency.40 ilar to that of deferoxamine.44,48 Deferasirox is
Most of these effects can be monitored and con- administered once daily and appears to have an
trolled. acceptable side-effect profile.44,49,50 Toxic effects
Deferiprone has a number of advantages over that have been observed have been related mainly
deferoxamine. It can penetrate the cell membrane to iron deprivation and transient gastrointestinal
and chelate toxic intracellular iron species.41 In a symptoms. No cases of agranulocytosis have been
preliminary study, hemoglobin levels increased and reported in several phase 2 trials involving several
transfusion requirements decreased in a few patients hundred patients.49,50
with hemoglobin E thalassemia who were treated In summary, a growing body of evidence sug-
with deferiprone for an average of 50 weeks.42 Most gests that deferiprone is an acceptable alternative
important, recent evidence suggests that defer- for patients who are unable to receive deferoxa-

1140 n engl j med 353;11 www.nejm.org september 15 , 2005

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

mine.33 The combination of deferiprone and de- selection, and the development of alternative sourc-
feroxamine appears very promising but requires es of hematopoietic stem cells.
further verification. The preliminary data on de- Low-risk patients (those with thalassemia termed
ferasirox are encouraging, and long-term clinical class 1 or class 2 by the Lucarelli classification, which
trials are still required. Finally, improved noninva- is used to assess risk factors that predict outcome
sive technologies (including imaging and blood and prognosis and addresses the degree of hepa-
tests) for quantitation of iron overload will provide tomegaly, the presence of portal fibrosis on liver
reliable information for the assessment of the effi- biopsy, and the effectiveness of chelation therapy
cacy of present and future therapies. before transplantation) have had excellent results
after bone marrow transplantation; however, pa-
hypercoagulability tients with class 3 disease (with extensive liver dam-
age from iron overload) have had poor results in
Thromboembolic phenomena, both venous and the past, primarily because of the 30 percent rate of
arterial, are not uncommon in patients with thalas- graft rejection due to attenuated conditioning.54 A
semia, particularly in patients who have undergone new preparatory regimen (including hydroxyurea,
splenectomy and who undergo transfusion infre- azathioprine, fludarabine, busulfan, and cyclophos-
quently.51 Abnormalities in the levels of coagula- phamide) has substantially improved the results in
tion factors and their inhibitors have been report- patients with class 3 disease who are younger than
ed, resulting in what can be defined as a chronic 17 years of age. The survival rate among these pa-
hypercoagulable state.16 tients was 93 percent, and the rejection rate fell to
Erythrocyte-membrane abnormalities contrib- 8 percent.55
ute to hypercoagulability (Fig. 2). Membrane lipid Because of the difficulty of eradicating the en-
peroxidation increases the surface expression of an- dogenous thalassemic bone marrow, it has been
ionic phospholipids such as phosphatidylserine.14 considered essential to administer full myeloab-
Exposure of phosphatidylserine on the erythrocyte lative conditioning regimens for transplantation.
was highly correlated with the expression of plate- Nonmyeloablative regimens have rarely been at-
let activation markers.16 Erythrocytes that are ex- tempted.56,57 Follow-up is short, and it is unclear
posed to phosphatidylserine may also contribute whether this approach will be beneficial. However,
directly to the vascular damage observed in thalas- in contrast to what may occur after bone marrow
semia.52 In addition, erythrocytes and platelets from transplantation for cancer, long-term mixed chi-
patients with thalassemia contain higher levels of merism can sometimes result in an acceptable clin-
reactive oxygen species and lower levels of intracel- ical outcome in thalassemia as long as the anemia
lular glutathione than do normal erythrocytes and is corrected.58
platelets, and this finding may be attributed to con- An increase in the available donor pool for bone
tinuous exposure to oxidative insults.53 Further marrow transplantation has been achieved by us-
studies will be required before conclusive recom- ing matched, unrelated donors.59 Extended haplo-
mendations can be made for prophylactic antico- typing has been developed for unrelated donors,
agulation, antiplatelet therapy, or both for patients and the resulting outcomes were similar to those
with thalassemia who are at risk (during pregnancy using matched, related donors.60 The use of relat-
or in the postoperative period) or on a routine basis, ed or unrelated umbilical-cord blood further in-
particularly in patients who have undergone sple- creases the donor pool. However, cord-blood trans-
nectomy. plantations have often been unsuccessful in the
treatment of thalassemia because large numbers of
transplanted cells need to be administered to sus-
hematopoietic stem- cell
transplantation tain hematopoiesis and prevent graft rejection. In
one study using related cord blood,61 7 of 33 pedi-
Although hematopoietic stem-cell transplantation atric patients rejected grafts and 3 others exhibited
is the only available curative approach for thalasse- stable mixed chimerism. Another study reported a
mia, it has been limited by the high cost and the high rate of nonengraftment and secondary rejec-
scarcity of HLA-matched, related donors. The past tion: only four of nine children were transfusion-
several years have brought progress in the realms independent at a median follow-up period of 49
of conditioning regimens, donor identification and months.62 Rare case reports have described a suc-

n engl j med 353;11 www.nejm.org september 15, 2005 1141

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

cessful outcome with unrelated cord-blood trans- erythropoiesis” without raising fetal hemoglobin.
plantation.63,64 In the future, umbilical-cord-blood The effect appeared to be dose-dependent and was
transplantation may be more successful if stem cells observed primarily in patients with b-thalassemia
can be expanded ex vivo. intermedia who had undergone splenectomy.71 Re-
In summary, hematopoietic stem-cell transplan- cently, long-acting darbepoetin alfa was shown to
tation with the use of related or unrelated donors increase hemoglobin levels substantially in patients
is a viable alternative that generally results in an ex-
with hemoglobin E–b-thalassemia disease.72 Two
cellent outcome for low-risk patients. If transplan- important obstacles to the use of recombinant hu-
tation is successful, transfusions, and usually chela- man erythropoietin are its relatively high cost and its
tion therapy, are no longer necessary. There is a subcutaneous administration route, which restrict
small risk of serious complications (death, graft its use in developing countries. Appropriate clinical
failure or rejection, and graft-versus-host disease). protocols are needed to delineate the role of recom-
Furthermore, growth failure and endocrinopathies, binant human erythropoietin (alone or in combina-
particularly gonadal dysfunction, can still occur.23,65tion with the aforementioned drugs) in the treat-
All these factors, in addition to the availability of ad-
ment of b-thalassemia.
equate supportive care in various geographical re- Screening for new compounds that augment fe-
gions, must be considered when deciding whether tal hemoglobin production can be performed with
to perform transplantation in any given patient. the use of cell-culture techniques, and various ani-
mal models have been useful in the evaluation of
experimental therapies potential fetal hemoglobin inducers.73 Further-
more, perturbations of growth-related signaling
cellular and molecular modifiers appear to activate tissue-specific fetal genes.74 Ex-
Augmenting the synthesis of fetal hemoglobin ploration of these signaling pathways may lead to
should ameliorate the severity of b-thalassemia.1 the discovery of new pharmacologic agents for the
Drugs such as 5-azacytidine, hydroxyurea, and var- treatment of thalassemia.
ious butyrate derivatives have been used for this pur-
pose.66 Hydroxyurea has shown substantial ben- potential role of antioxidants
efits in a subgroup of patients with sickle cell Because reactive oxygen species play an important
anemia67 but has been used less often in thalasse- role in the pathophysiology of thalassemia9 (Fig. 2),
mia. In a few pediatric patients with thalassemia, antioxidants may be an effective therapy. Patients
transfusion requirements were eliminated after with thalassemia have very high plasma levels of
treatment with hydroxyurea for approximately 20 malonyldialdehyde, a by-product of lipid peroxida-
months.68 In general, preliminary results among a tion. Malonyldialdehyde levels correlate positively
relatively small number of patients have been incon- with serum iron and with non–transferrin-bound
sistent, and thus the role of hydroxyurea in thalas- iron.75 Elevated levels of reactive oxygen species
semia therapy remains uncertain.66 tended to normalize in response to oral therapy with
One possible explanation for the differential ef- vitamin E, with patients exhibiting improvement in
fects of hydroxyurea in sickle cell anemia as com- the antioxidant–oxidant balance in plasma and
pared with thalassemia is that many patients with decreased lipid peroxidation in erythrocytes.76 Plant
thalassemia are transfusion-dependent. Hypertrans- flavonoids (including rutin and curcumin) are an-
fusion suppresses endogenous erythropoiesis, par- other group of antioxidants with therapeutic po-
ticularly of hydroxyurea-responsive types of cells.67 tential in thalassemia. These compounds have salu-
Therefore, despite the beneficial effects of hy- tary effects on erythrocytes that have been damaged
droxyurea on erythropoiesis,66 it will be difficult to by oxidation. Polyphenols (a major component of
correct the anemia associated with thalassemia with tea) bind to ferric iron and could also protect thal-
the use of hydroxyurea. Moreover, genetic predis- assemic erythrocytes from oxidation.77 However,
position, such as the presence of Xmnl polymor- despite their apparent salutary effects on erythro-
phism,69 and the type of thalassemia, such as hemo- cytes, antioxidants have not yet been shown to ame-
globin E thalassemia,70 may determine the response liorate the anemia of patients with thalassemia.
to hydroxyurea treatment. Antioxidants may be more effective if used in
Recombinant human erythropoietin was shown combination — for example, as a lipid antioxidant
to provide the benefit of increasing “thalassemic like vitamin E, together with N-acetylcysteine, which

1142 n engl j med 353;11 www.nejm.org september 15, 2005

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

is a protein antioxidant that improves several mea- sion. Small interfering RNA corresponding to tran-
sures in oxidized sickle erythrocytes,78 and an iron scripts of BP1 (a protein that negatively regulates
chelator such as deferiprone. This approach, if b-globin expression by binding to its upstream re-
successful, could be particularly useful in countries gion) enhanced b-globin promoter activity in eryth-
with limited financial resources. roid cells.90
Another new molecular approach uses so-called
experimental molecular therapies antisense technology for correction of the molec-
Initial efforts at gene therapy were directed against ular defect caused by the thalassemia mutation. Syn-
diseases of the b-globin gene. This therapeutic thesis of hemoglobin A was restored and alternate
strategy involves the insertion of a normally func- splicing corrected with the use of antisense oligo-
tioning g-globin or b-globin gene into the patient’s nucleotides that blocked splicing at the mutant
autologous hematopoietic stem cells. Although the alternate splice site, thereby forcing the splicing
concept is relatively straightforward, it has met with machinery to select the normal splice site.91 In addi-
two decades of seemingly insurmountable hurdles tion, lentiviral vectors carrying an altered U7 small
that have been well summarized elsewhere.79,80 nuclear RNA gene encoding an antisense RNA have
The major problems with this type of gene thera- been shown to correct splicing defects caused by
py have been related to vector construction. The ge- thalassemia mutations.92
netic elements of the vector that are necessary for ap- Clearly, these types of therapies remain highly
propriate regulation of the inserted gene have been experimental; accordingly, their clinical potential
defined.79 However, the therapeutic gene must be remains uncertain. Nevertheless, these methods
inserted into a hematopoietic stem cell and must be may be useful molecular approaches for the devel-
expressed at high levels, over an extended period, in opment of new therapies for thalassemias.
an erythroid-specific manner. In addition, the vector
must be safe from recombination or mutagenesis. progress in prenatal diagnosis
Oncoretroviral and adenoviral vectors have been
found to be unsuitable for various reasons.80 Polymerase-chain-reaction (PCR) technology has
The introduction of lentiviral vectors was an been used for more than a decade to detect point
important advance, since these viruses do not re- mutations or deletions in chorionic-villus samples,
quire cell division for entry into eukaryotic cells enabling first-trimester, DNA-based testing for thal-
and can stably hold larger DNA inserts without re- assemia. However, because pregnancy termination
arrangements.81,82 Self-inactivating lentiviral vec- is unacceptable to some persons (even when the
tors were constructed to address safety issues.83 fetus is affected), methods were developed, begin-
Longer locus-control-region elements84 may abro- ning in the early 1990s, to perform diagnostic test-
gate position effects that reduce expression of the ing before implantation. Preimplantation genetic
therapeutic gene. The problem of gene silencing of diagnosis93 involves performing conventional in
the transduced gene85 has been approached by the vitro fertilization, followed by removal of one or
use of insulators, which are DNA sequences that two cells from the resulting blastomeres on day 3.
are thought to function as boundary elements pro- PCR is then used to detect thalassemia mutations
tecting against chromatin-dependent repression within the cells that have been removed so that un-
of genetic activity.86,87 A lentiviral vector carrying affected blastomeres may be selected for implanta-
both the b-globin gene and an insulator stably cor- tion. Preimplantation genetic diagnosis requires a
rected the b-thalassemia phenotype in human b- high degree of technical expertise. Furthermore,
thalassemia progenitor cells that were transplant- the phenomenon of “allele dropout” (failure to am-
ed into immune-deficient mice.88 However, a re- plify one of the two alleles in a heterozygous cell)
cent report demonstrated that lentiviral constructs can result in diagnostic errors.94 Nonetheless, this
preferentially integrated at intragenic sites, often technology has been successful, and improvements
within genes that regulate hematopoiesis.89 Be- in outcome have led to its use in many countries.
sides being associated with lower expression lev- Recently, preimplantation genetic diagnosis has
els,87 intragenic integration raises new concerns been extended to HLA typing on embryonic biop-
regarding the safety of these vectors.89 sies, which allows the selection of an embryo that
Small interfering RNA is the basis for a new is not affected by thalassemia and that may also
strategy to augment transduced b-globin expres- serve as a stem-cell donor for a previously affected

n engl j med 353;11 www.nejm.org september 15, 2005 1143

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

child within the same family. Recent reports95,96 countries emphasize curative forms of therapy, such
have confirmed that this approach is feasible. How- as bone marrow transplantation and gene therapy,
ever, serious ethical concerns have been raised.93 which require compliant patients with access to the
Although it is considered ethical not to implant an newest medications and sophisticated scientific fa-
embryo that is affected with a serious genetic dis- cilities. Western cultures need to develop improved
order, in certain countries it is forbidden to select support for patients with thalassemia and their fam-
an embryo on the basis of its designated role as a ilies. Doing so will prevent psychosocial issues from
potential stem-cell donor. taking their heavy toll due to noncompliance.100
Future prenatal diagnosis may be performed In contrast, the treatment of thalassemia is en-
noninvasively, with the use of maternal blood sam- tirely different in less developed countries, where
ples to isolate either fetal cells97 or fetal DNA98 for most of the patients with this disease reside. Safe
analysis. These techniques are feasible but have transfusion (with the use of filtration and the viral
not yet been perfected. Furthermore, techniques testing of blood) and chelation are not universally
using DNA in maternal plasma to exclude thalas- available. Consequently, many patients with thal-
semia in the fetus are applicable only to couples assemia in underdeveloped nations die in child-
for whom the paternal and maternal mutations hood or adolescence. Programs that provide ac-
are different. ceptable care, including transfusion of safe blood
and supportive therapy including chelation, must
be established. Thalassemia-prevention protocols
treatment in the developed must be developed in these countries, with the use
versus the underdeveloped
world of better education and screening and improved
access to prenatal diagnosis.101,102 The challenge
In developed parts of the world, such as the United for the future is to ensure that people who are
States and Europe, there are approximately 10,000 born with a severe form of thalassemia will con-
homozygous patients with thalassemia, and the tinue to thrive, while effective prevention eventu-
number of new cases has been progressively de- ally decreases the number of severely affected pa-
creasing because of effective prevention methods.99 tients worldwide.
Comprehensive, high-quality medical care is avail- We are indebted to Drs. Griffin Rodgers and Eitan Fibach for their
able in these countries, with longer life expectancy critical reading of the manuscript.
and a relatively good quality of life.27,37 Western

refer enc es
1. Higgs DR, Thein SL, Woods WG. The tein impairs erythropoiesis and exacerbates tein defects in alpha and beta thalassemia.
molecular pathology of the thalassaemias. b-thalassemia. J Clin Invest 2004;114:1457- J Clin Invest 1989;83:404-10.
In: Weatherall DJ, Clegg B, eds. The thalas- 66. 13. Hershko C, Graham G, Bates GW, Rach-
saemia syndromes. 4th ed. Oxford, England: 8. Pootrakul P, Sirankapracha P, Hemsor- milewitz EA. Non-specific serum iron in thal-
Blackwell Science, 2001:133-91. ach S, et al. A correlation of erythrokinetics, assemia: an abnormal serum iron fraction of
2. Cunningham MJ, Macklin EA, Neufeld ineffective erythropoiesis, and erythroid pre- potential toxicity. Br J Haematol 1978;40:
EJ, Cohen AR. Complications of b-thalasse- cursor apoptosis in Thai patients with thal- 255-63.
mia major in North America. Blood 2004; assemia. Blood 2000;96:2606-12. 14. Kuypers FA, de Jong K. The role of phos-
104:34-9. 9. Rachmilewitz EA, Schrier S. The patho- phatidylserine in recognition and removal
3. Old JM, Olivieri NF, Thein SL. Diagnosis physiology of b-thalassemia. In: Steinberg of erythrocytes. Cell Mol Biol 2004;50:147-
and management of thalassaemia. In: Weath- MH, Forget BG, Higgs DR, Nagel RL, eds. 58.
erall DJ, Clegg B, eds. The thalassaemia syn- Disorders of hemoglobin: genetics, patho- 15. Tavazzi D, Duca L, Graziadei G, Comino
dromes. 4th ed. Oxford, England: Blackwell physiology, and clinical management. Cam- A, Fiorelli G, Cappellini MD. Membrane-
Science, 2001:630-85. bridge, England: Cambridge University Press, bound iron contributes to oxidative damage
4. Karnon J, Zeuner D, Brown J, Ades AE, 2001:233-51. of b-thalassemia intermedia erythrocytes.
Wonke B, Modell B. Lifetime treatment costs 10. Advani R, Sorenson S, Shinar E, Lande Br J Haematol 2001;112:48-50.
of b-thalassaemia major. Clin Lab Haematol W, Rachmilewitz E, Schrier SL. Character- 16. Eldor A, Rachmilewitz EA. The hyperco-
1999;21:377-85. ization and comparison of the red blood cell agulable state in thalassemia. Blood 2002;
5. Angastiniotis M, Modell B. Global epi- membrane damage in severe human a- and 99:36-43.
demiology of hemoglobin disorders. Ann b-thalassemia. Blood 1992;79:1058-63. 17. Centis F, Tabellini L, Lucarelli G, et al.
N Y Acad Sci 1998;850:251-9. 11. Higgs DR, Thein SL, Wood WG. The The importance of erythroid expansion in
6. Weatherall DJ. Phenotype-genotype rela- pathophysiology of the thalassaemias. In: determining the extent of apoptosis in eryth-
tionships in monogenic disease: lessons Weatherall DJ, Clegg B, eds. The thalassaemia roid precursors in patients with b-thalasse-
from the thalassaemias. Nat Rev Genet 2001; syndromes. 4th ed. Oxford, England: Black- mia major. Blood 2000;96:3624-9.
2:245-55. well Science, 2001:192-236. 18. Mathias LA, Fisher TC, Zeng L, et al.
7. Kong Y, Zhou S, Kihm AJ, et al. 12. Shinar E, Rachmilewitz EA, Lux SE. Dif- Ineffective erythropoiesis in b-thalassemia
Loss of alpha-hemoglobin-stabilizing pro- fering erythrocyte membrane skeletal pro- major is due to apoptosis at the polychro-

1144 n engl j med 353;11 www.nejm.org september 15, 2005

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

matophilic normoblast stage. Exp Hematol 36. Esposito BP, Breuer W, Sirankapracha P, due to b-thalassemia. J Clin Pharmacol 2003;
2000;28:1343-53. Pootrakul P, Hershko C, Cabantchik ZI. Labile 43:565-72.
19. De Maria R, Testa U, Luchetti L, et al. plasma iron in iron overload: redox activity 50. Nisbet-Brown E, Olivieri NF, Giardina
Apoptotic role of Fas/Fas ligand system in and susceptibility to chelation. Blood 2003; PJ, et al. Effectiveness and safety of ICL670
the regulation of erythropoiesis. Blood 1999; 102:2670-7. in iron-loaded patients with thalassaemia:
93:796-803. 37. Borgna-Pignatti C, Rugolotto S, De Ste- a randomised, double-blind, placebo-con-
20. Testa U. Apoptotic mechanisms in the fano P, et al. Survival and complications in trolled, dose-escalation trial. Lancet 2003;
control of erythropoiesis. Leukemia 2004; patients with thalassemia major treated with 361:1597-602.
18:1176-99. transfusion and deferoxamine. Haemato- 51. Cappellini MD, Robbiolo L, Bottasso
21. Angelucci E, Bai H, Centis F, et al. logica 2004;89:1187-93. BM, Coppola R, Fiorelli G, Mannucci AP.
Enhanced macrophagic attack on b-thalas- 38. Franchini M, Veneri D. Iron-chelation Venous thromboembolism and hypercoag-
semia major erythroid precursors. Haema- therapy: an update. Hematol J 2004;5:287- ulability in splenectomized patients with
tologica 2002;87:578-83. 92. thalassaemia intermedia. Br J Haematol
22. Dzik WH. Leukoreduction of blood 39. Wanless IR, Sweeney G, Dhillon AP, et 2000;111:467-73.
components. Curr Opin Hematol 2002;9: al. Lack of progressive hepatic fibrosis dur- 52. Manodori AB, Barabino GA, Lubin BH,
521-6. ing long-term therapy with deferiprone in Kuypers FA. Adherence of phosphatidyl-
23. De Sanctis V. Growth and puberty and subjects with transfusion-dependent b-thal- serine-exposing erythrocytes to endothelial
its management in thalassaemia. Horm Res assemia. Blood 2002;100:1566-9. [Erratum, matrix thrombospondin. Blood 2000;95:
2002;58:Suppl 1:72-9. Blood 2003;101:2460.] 1293-300.
24. Raiola G, Galati MC, De Sanctis V, et al. 40. Ceci A, Baiardi P, Felisi M, et al. The 53. Amer J, Fibach E. Oxidative status of
Growth and puberty in thalassemia major. safety and effectiveness of deferiprone in a platelets in normal and thalassemic blood.
J Pediatr Endocrinol Metab 2003;16:Suppl large-scale, 3-year study in Italian patients. Thromb Haemost 2004;92:1052-9.
2:259-66. Br J Haematol 2002;118:330-6. 54. Lucarelli G, Clift RA, Galimberti M, et
25. Cohen AR, Galanello R, Pennell DJ, 41. Shalev O, Repka T, Goldfarb A, et al. al. Marrow transplantation for patients with
Cunningham MJ, Vichinsky E. Thalassemia. Deferiprone (L1) chelates pathologic iron thalassemia: results in class 3 patients.
Hematology (Am Soc Hematol Educ Pro- deposits from membranes of intact thalas- Blood 1996;87:2082-8.
gram) 2004;1:14-34. semic and sickle red blood cells both in vitro 55. Sodani P, Gaziev D, Polchi P, et al. New
26. Voskaridou E, Terpos E. New insights and in vivo. Blood 1995;86:2008-13. approach for bone marrow transplanta-
into the pathophysiology and management 42. Pootrakul P, Sirankapracha P, Sankote J, tion in patients with class 3 thalassemia
of osteoporosis in patients with beta thalas- et al. Clinical trial of deferiprone iron chela- aged younger than 17 years. Blood 2004;
saemia. Br J Haematol 2004;127:127-39. tion therapy in b-thalassaemia/haemoglo- 104:1201-3.
27. Modell B, Khan M, Darlison M. Survival bin E patients in Thailand. Br J Haematol 56. Iannone R, Casella JF, Fuchs EJ, et al.
in beta-thalassaemia major in the UK: data 2003;122:305-10. Results of minimally toxic nonmyeloabla-
from the UK Thalassaemia Register. Lancet 43. Piga A, Gaglioti C, Fogliacco E, Tricta F. tive transplantation in patients with sickle
2000;355:2051-2. Comparative effects of deferiprone and de- cell anemia and beta-thalassemia. Biol Blood
28. Olivieri NF. The b-thalassemias. N Engl feroxamine on survival and cardiac disease Marrow Transplant 2003;9:519-28.
J Med 1999;341:99-109. [Erratum, N Engl J in patients with thalassemia major: a retro- 57. Hongeng S, Chuansumrit A, Hathirat P,
Med 1999;341:1407.] spective analysis. Haematologica 2003;88: Rerkamnuaychoke B, Chaisiripoomkere W,
29. Ganz T. Hepcidin, a key regulator of 489-96. Jootar S. Full chimerism in nonmyeloabla-
iron metabolism and mediator of anemia of 44. Hershko C, Link GM, Konijn AM, tive stem cell transplantation in a beta-thal-
inflammation. Blood 2003;102:783-8. Cabantchik ZI. Iron chelation therapy. Curr assemia major patient (class 3 Lucarelli).
30. Papanikolaou G, Tzilianos M, Christakis Hematol Rep 2005;4:110-6. Bone Marrow Transplant 2002;30:409-10.
JI, et al. Hepcidin in iron overload disorders. 45. Giardina PJ, Grady RW. Chelation ther- 58. Andreani M, Nesci S, Lucarelli G, et al.
Blood 2005;105:4103-5. apy in b thalassemia: an optimistic update. Long-term survival of ex-thalassemic patients
31. Rachmilewitz EA, Weizer-Stern O, Adam- Semin Hematol 2001;38:360-6. with persistent mixed chimerism after bone
sky K, et al. Iron and oxidative stress in thal- 46. Link G, Konijn AM, Breuer W, Cabantchik marrow transplantation. Bone Marrow Trans-
assemia: Eighth International Cooley’s Ane- ZI, Hershko C. Exploring the “iron shuttle” plant 2000;25:401-4.
mia Symposium, March 2005. Ann N Y Acad hypothesis in chelation therapy: effects of 59. Krishnamurti L, Abel S, Maiers M,
Sci 2005;1054:1-6. combined deferoxamine and deferiprone Flesch S. Availability of unrelated donors for
32. Hoffbrand AV, Cohen A, Hershko C. treatment in hypertransfused rats with hematopoietic stem cell transplantation for
Role of deferiprone in chelation therapy for labeled iron stores and in iron-loaded rat hemoglobinopathies. Bone Marrow Trans-
transfusional iron overload. Blood 2003;102: heart cells in culture. J Lab Clin Med 2001; plant 2003;31:547-50.
17-24. 138:130-8. 60. La Nasa G, Giardini C, Argiolu F, et al.
33. Brittenham GM, Cohen AR, McLaren 47. Mourad FH, Hoffbrand AV, Sheikh-Taha Unrelated donor bone marrow transplanta-
CE, et al. Hepatic iron stores and plasma fer- M, Koussa S, Khoriaty AI, Taher A. Compar- tion for thalassemia: the effect of extended
ritin concentration in patients with sickle ison between desferrioxamine and com- haplotypes. Blood 2002;99:4350-6.
cell anemia and thalassemia major. Am J bined therapy with desferrioxamine and 61. Locatelli F, Rocha V, Reed W, et al.
Hematol 1993;42:81-5. deferiprone in iron overloaded thalassaemia Related umbilical cord blood transplanta-
34. Anderson LJ, Wonke B, Prescott E, patients. Br J Haematol 2003;121:187-9. tion in patients with thalassemia and sickle
Holden S, Walker JM, Pennell DJ. Compar- 48. Cappellini M, Bejaoui M, Perrotta S, et cell disease. Blood 2003;101:2137-43.
ison of effects of oral deferiprone and sub- al. Phase III evaluation of once-daily, oral 62. Fang J, Huang S, Chen C, et al. Umbili-
cutaneous desferrioxamine on myocardial therapy with ICL670 (Exjade) versus defer- cal cord blood transplantation in Chinese
iron concentrations and ventricular func- oxamine in patients with b-thalassemia and children with beta-thalassemia. J Pediatr
tion in beta-thalassemia. Lancet 2002;360: transfusional hemosiderosis. Blood 2004; Hematol Oncol 2004;26:185-9.
516-20. 104:984a. abstract. 63. Hall JG, Martin PL, Wood S, Kurtzberg J.
35. Voskaridou E, Douskou M, Terpos E, et 49. Galanello R, Piga A, Alberti D, Rouan Unrelated umbilical cord blood transplanta-
al. Magnetic resonance imaging in the eval- MC, Bigler H, Sechaud R. Safety, tolerability, tion for an infant with beta-thalassemia major.
uation of iron overload in patients with and pharmacokinetics of ICL670, a new J Pediatr Hematol Oncol 2004;26:382-5.
b thalassemia and sickle cell disease. Br J orally active iron-chelating agent in patients 64. Tan PH, Hwang WY, Goh YT, et al. Unre-
Haematol 2004;126:736-42. with transfusion-dependent iron overload lated peripheral blood and cord blood

n engl j med 353;11 www.nejm.org september 15, 2005 1145

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.
medical progress

hematopoietic stem cell transplants for thal- 77. Rund D, Rachmilewitz EA. Pathophysi- globin promoter activity and beta-globin
assemia major. Am J Hematol 2004;75:209- ology of a- and b-thalassemia: therapeutic mRNA levels in the human erythroleukemia
12. implications. Semin Hematol 2001;38:343- (K562) cell line. Exp Hematol 2004;32:700-
65. Li CK, Chik KW, Wong GW, Cheng PS, 9. 8.
Lee V, Shing MM. Growth and endocrine 78. Pace BS, Shartava A, Pack-Mabien A, 91. Lacerra G, Sierakowska H, Carestia C, et
function following bone marrow transplan- Mulekar M, Ardia A, Goodman SR. Effects al. Restoration of hemoglobin A synthesis
tation for thalassemia major. Pediatr Hema- of N-acetylcysteine on dense cell formation in erythroid cells from peripheral blood of
tol Oncol 2004;21:411-9. in sickle cell disease. Am J Hematol 2003; thalassemic patients. Proc Natl Acad Sci
66. Atweh GF, Loukopoulos D. Pharmaco- 73:26-32. U S A 2000;97:9591-6.
logical induction of fetal hemoglobin in 79. Persons DA, Nienhuis AW. Gene ther- 92. Vacek MM, Ma H, Gemignani F, Lacerra
sickle cell disease and b-thalassemia. Semin apy for the hemoglobin disorders. Curr G, Kafri T, Kole R. High-level expression of
Hematol 2001;38:367-73. Hematol Rep 2003;2:348-55. hemoglobin A in human thalassemic eryth-
67. Steinberg MH, Barton F, Castro O, et al. 80. Puthenveetil G, Malik P. Gene therapy roid progenitor cells following lentiviral vec-
Effect of hydroxyurea on mortality and mor- for hemoglobinopathies: are we there yet? tor delivery of an antisense snRNA. Blood
bidity in adult sickle cell anemia: risks and Curr Hematol Rep 2004;3:298-305. 2003;101:104-11.
benefits up to 9 years of treatment. JAMA 81. May C, Rivella S, Callegari J, et al. Ther- 93. Braude P, Pickering S, Flinter F, Ogilvie
2003;289:1645-51. [Erratum, JAMA 2003; apeutic haemoglobin synthesis in beta- CM. Preimplantation genetic diagnosis. Nat
290:756.] thalassaemic mice expressing lentivirus- Rev Genet 2002;3:941-53. [Erratum, Nat
68. Bradai M, Abad MT, Pissard S, Lamraoui encoded human beta-globin. Nature 2000; Rev Genet 2003;4:157.]
F, Skopinski L, de Montalembert M. Hydrox- 406:82-6. 94. Piyamongkol W, Bermudez MG, Harper
yurea can eliminate transfusion require- 82. Rivella S, Sadelain M. Therapeutic globin JC, Wells D. Detailed investigation of factors
ments in children with severe b-thalasse- gene delivery using lentiviral vectors. Curr influencing amplification efficiency and
mia. Blood 2003;102:1529-30. Opin Mol Ther 2002;4:505-14. allele drop-out in single cell PCR: implica-
69. Gilman JG, Huisman TH. DNA sequence 83. Moreau-Gaudry F, Xia P, Jiang G, et al. tions for preimplantation genetic diagnosis.
variation associated with elevated fetal G High-level erythroid-specific gene expres- Mol Hum Reprod 2003;9:411-20.
gamma globin production. Blood 1985;66: sion in primary human and murine hemato- 95. Fiorentino F, Biricik A, Karadayi H, et al.
783-7. poietic cells with self-inactivating lentiviral Development and clinical application of a
70. Fucharoen S, Siritanaratkul N, Winicha- vectors. Blood 2001;98:2664-72. strategy for preimplantation genetic diagno-
goon P, et al. Hydroxyurea increases hemo- 84. Hanawa H, Hargrove PW, Kepes S, sis of single gene disorders combined with
globin F levels and improves the effectiveness Srivastava DK, Nienhuis AW, Persons DA. HLA matching. Mol Hum Reprod 2004;10:
of erythropoiesis in b-thalassemia/hemoglo- Extended beta-globin locus control region 445-60.
bin E disease. Blood 1996;87:887-92. elements promote consistent therapeutic 96. Van de Velde H, Georgiou I, De Rycke M,
71. Rachmilewitz EA, Aker M, Perry D, Dover expression of a gamma-globin lentiviral vec- et al. Novel universal approach for preim-
G. Sustained increase in haemoglobin and tor in murine beta-thalassemia. Blood 2004; plantation genetic diagnosis of beta-thalas-
RBC following long-term administration of 104:2281-90. saemia in combination with HLA matching
recombinant human erythropoietin to patients 85. Lung HY, Meeus IS, Weinberg RS, Atweh of embryos. Hum Reprod 2004;19:700-8.
with homozygous b-thalassaemia. Br J Hae- GF. In vivo silencing of the human gamma- 97. Di Naro E, Ghezzi F, Vitucci A, et al. Pre-
matol 1995;90:341-5. globin gene in murine erythroid cells fol- natal diagnosis of b-thalassemia using fetal
72. Singer ST, Sweeters N, Vichinsky EP, lowing retroviral transduction. Blood Cells erythroblasts enriched from maternal blood
Wagner AJ, Rachmilewitz EA. A dose-find- Mol Dis 2000;26:613-9. by a novel gradient. Mol Hum Reprod 2000;
ing and safety study of darbepoietin alfa 86. Rivella S, Callegari JA, May C, Tan CW, 6:571-4.
(erythropoiesis stimulating protein) for the Sadelain M. The cHS4 insulator increases 98. Ding C, Chiu RWK, Lau TK, et al. MS
treatment of anemia in patients with thalas- the probability of retroviral expression at ran- analysis of single-nucleotide differences in
semia intermedia. Blood 2003;102:268a. dom chromosomal integration sites. J Virol circulating nucleic acids: application to non-
abstract. 2000;74:4679-87. invasive prenatal diagnosis. Proc Natl Acad
73. Fibach E. Cell culture and animal mod- 87. Emery DW, Yannaki E, Tubb J, Nishino Sci U S A 2004;101:10762-7.
els to screen for promising fetal hemoglo- T, Li Q, Stamatoyannopoulos G. Develop- 99. Cao A, Galanello R. Effect of consan-
bin-stimulating compounds. Semin Hema- ment of virus vectors for gene therapy of guinity on screening for thalassemia. N Engl
tol 2001;38:374-81. beta chain hemoglobinopathies: flanking J Med 2002;347:1200-2.
74. Bhanu NV, Trice TA, Lee YT, Miller JL. with a chromatin insulator reduces gamma- 100. Anie KA, Massaglia P. Psychological
A signaling mechanism for growth-related globin gene silencing in vivo. Blood 2002; therapies for thalassaemia. Cochrane Data-
expression of fetal hemoglobin. Blood 2004; 100:2012-9. base Syst Rev 2001;3:CD002890.
103:1929-33. 88. Puthenveetil G, Scholes J, Carbonell D, 101. Ahmed S, Saleem M, Modell B, Petrou
75. Cighetti G, Duca L, Bortone L, et al. et al. Successful correction of the human M. Screening extended families for genetic
Oxidative status and malondialdehyde in beta-thalassemia major phenotype using a hemoglobin disorders in Pakistan. N Engl J
b-thalassemia patients. Eur J Clin Invest lentiviral vector. Blood 2004;104:3445-53. Med 2002;347:1162-8.
2002;32:Suppl 1:55-60. 89. Imren S, Fabry ME, Westerman KA, et 102. Alwan A, Modell B. Recommenda-
76. Tesoriere L, D’Arpa D, Butera D, et al. al. High-level beta-globin expression and tions for introducing genetics services in
Oral supplements of vitamin E improve mea- preferred intragenic integration after len- developing countries. Nat Rev Genet 2003;
sures of oxidative stress in plasma and reduce tiviral transduction of human cord blood 4:61-8.
oxidative damage to LDL and erythrocytes stem cells. J Clin Invest 2004;114:953-62. Copyright © 2005 Massachusetts Medical Society.
in b-thalassemia intermedia patients. Free 90. Zoueva OP, Rodgers GP. Inhibition of
Radic Res 2001;34:529-40. beta protein 1 expression enhances beta-

1146 n engl j med 353;11 www.nejm.org september 15, 2005

Downloaded from www.nejm.org on April 10, 2007 . Copyright © 2005 Massachusetts Medical Society. All rights reserved.

Anda mungkin juga menyukai