Anda di halaman 1dari 100

End-to-End Single-Use

Process for Monoclonal


Antibody Manufacturing
e Adaptable Laboratory
Raman Spectroscopy Applied
to Pharmaceutical Tablets
Amorphous Solid Dispersions
Approaches to Quantication
by Powder X-ray Diraction w
w
w
.
a
m
e
r
i
c
a
n
p
h
a
r
m
a
c
e
u
t
i
c
a
l
r
e
v
i
e
w
.
c
o
m
American Pharmaceutical
The Review of American Pharmaceutical Business & Technology
Volume 14 Issue 1 | Jan/Feb 2011
A
m
e
r
i
c
a
n

P
h
a
r
m
a
c
e
u
t
i
c
a
l

R
e
v
i
e
w


|


V
o
l
u
m
e

1
4

I
s
s
u
e

1


|


J
a
n
/
F
e
b

2
0
1
1
SHOW ISSUE
FC_APRJanFeb2011.indd 1 2/11/11 3:53 PM
endo ad campaign 20102.indd 1 10/15/2010 12:10:01 PM
Xcelolab Powder Dispenser
Provides fast, exible, precision powder dispensing for any laboratory. Performs
accurate, closed loop, weight dispensing that is difcult and potentially inaccurate
by hand or by other laboratory methods.
Xcelodose S Precision Powder Micro-Dosing System
Precisely dispenses drug substances alone, as low as 100g, into capsules
without excipients or bulking agents. This allows companies to eliminate costly
and time-consuming compatibility, stability and preformulation studies during early
stage drug development.
CFS 1200 and the NEW CFS 1500 C Liquid Filling & Sealing Machines
These fully automatic cGMP compliant machines ll and seal liquids/semi-solids
into two-piece hard capsules at speeds of 1,200 and 1,500 capsules per hour.
This helps scientists exploit the potential of lipid-based formulations for poorly
soluble compounds.
CapsuleCaddy Container
The perfect complement to our R&D machines, it contains 10,000 Licaps,
DBcaps, Vcaps Plus or Coni-Snap capsules.
To receive a full R&D portfolio, call 888-783-6361
or send an email to marketing.AMER@pzer.com.
www.capsugel.com
Achieving Faster Time to First in Human

N
E
W
N
E
W
APR_19818_FIN_7-26-10.indd 1 7/26/10 11:37 AM
Editorial
EDITOR | Emily Johnson
emily.johnson@russpub.com
Sales
ACCOUNT MANAGER | Southeast | Joel Kern
joel.kern@russpub.com
INTERNATIONAL SALES MANAGER | Gary King
gary.king@russpub.com
REGIONAL SALES MANAGER | Laura Zoibi
laura.zoibi@russpub.com
Corporate
PRESIDENT | Nigel Russell
CIRCULATION DIRECTOR | Steve Koppelman
Production
ART DIRECTOR | Jennifer Campbell
PRODUCTION MANAGER | Brandon Waggoner
OFFICE MANAGER | Sandra Anwarzai
American Pharmaceutical Review
Published by

Russell Publishing L.L.C.
9225 Priority Way West Drive, Suite 120
Indianapolis, IN 46240-1467 USA
Tel: 317-816-8787 | Fax: 317-816-8788
www.russpub.com
E-mail: info@russpub.com
Website: www.americanpharmaceuticalreview.com
Printed in the USA
American Pharmaceutical Review (ISSN - 1099-8012) is pub-
lished seven times a year by Russell Publishing LLC, 9225 Priority
Way West Drive, Suite 120, Indianapolis, Indiana 46240-1467.
Subscription Rate (7 issues) 1 year $135.00. Periodicals postage
paid at Indianapolis, IN 46206 and additional mailing ofces.
POSTMASTER: Send change of addresses to Russell Publishing
LLC, 9225 Priority Way West Drive, Suite 120, Indianapolis,
Indiana 46240-1467. Publication Agreement #40739004. Return
Undeliverable Canadian Addresses to: Station A, PO Box 54,
Windsor ON N9A 6J5, E-mail: info@russpub.com
Copyright rests with the publishers.
All rights reserved 2010
Russell Publishing L.L.C.
No part of this publication may be reproduced, stored in a
retrieval system or transmitted in any form or by any means
electronic, mechanical, photocopying, recording or other-
wise without the prior written permission of the copyright
holder. While the publishers believe that all information
contained in this publication was correct at the time of going
to press, they can accept no liability for any inaccuracies
that may appear or loss sufered directly or indirectly by any
reader as a result of any advertisement, editorial, photo-
graphs or other material published in American Pharmaceuti-
cal Review.
2 |

|

January/February 2011
Dependable and Relevant
Your Source for Information in the
Pharmaceutical Industry
American Pharmaceutical Review,
Pharmaceutical Outsourcing, and
International Drug Discovery bring the most up
to date reviews, news, trends and comments
to an ever-changing industry. With their
complementary websites, e-newsletters and
webcasts they offer the complete spectrum
of marketing avenues for any company that
operates in the life science arena.
Amerian Pharmaceutical Review, Pharmaceutical Outsourcing,
and International Drug Discovery are products of Russell Publishing
9225 Priority Way West Drive, Indianapolis, IN 46240
317-816-8787 | Fax 317-816-8788
Harnessing the Power of Knowledge to Drive
World Class Science and Technology
April 11-15, 2011
JW Marriott San antonio Hill Country | San antonio, texaS
www.pda.org/annual2011
The Parenteral Drug Association presents...
1946
2011
65
TH
ANNIVERSARY
Meeting Apri l 11-13 | exhibition & Career Fair Apri l 11-12
PDa/FDa ProCess ValiDation guiDanCe Post ConFerenCe WorkshoP Apri l 13-14 | Courses Apri l 14-15
www.pda.org/annual2011
The Opening Keynote addresses include:
Join more than 50 speakers in over 45 presentations in sessions that will discuss:
Quality Science
Manufacturing/Process Science
Development Science
Outsourcing/Supply Chain
New in 2011 is the Fundamentals Track This track is designed for those who are new
to the pharmaceutical/biopharmaceutical industry or have recently changed jobs with a
change in focus.
Additional Educational and Training Opportunities!
Attend the PDA Process Validation Guidance Post Conference Workshop to learn about
US and international regulations, technology transfer, documentation strategies, post
approval reporting and more!
PDAs Training and Research Institute (PDA TRI) is offering seven courses in conjunction
with this meeting covering topics such as GMP regulations, Rapid Micro Methods, six
sigma in process validation, cleanroom management and more!
EMA Perspective: Knowledge Management Fostering Academic-Industry Collaboration for Product Development
M. Lynn Crimson,
Dean, School
of Pharmacy,
University of
Texas
Janet Walkow, PhD,
Director, Drug
Dynamics Institute,
University of Texas
Piotr Krauze, Scientifc
Administrator/Compliance
and Inspection Sector,
European Medicines
Agency
Ghulam Shabir Arain, PhD, FCQI
Principal Scientist
Abbott Laboratories, UK
Douglas J. Ball, M.S.
Diplomate, American Board of Toxicology; Research
Fellow, Drug Safety Research
& Development
Pfzer Global Research & Development
Suraj Baloda, Ph.D.
Director, QC Microbiology & Environmental Control
Ben Venue Laboratories, Inc.
Rory Budihandojo
Director, Quality Systems Audit
Boehringer Ingelheim Shanghai
Pharmaceuticals Co., Ltd.
Suggy S. Chrai, Ph.D., MBA., R.Ph.
President & CEO
Chrai Associates, Inc.
Melanie W. Cooke
Associate Scientist
Amgen, Inc.
Emil W. Ciurczak
Chief Technical Ofcer
Cadrai Technology Group
Rick E. Cooley
North America Manager of Process Analytics
Dionex Corporation
Walter Dziki, Ph.D.
Associate Research Fellow
Abbott Laboratories
John Finkbohner, Ph.D.
Director, Regulatory Afairs
MedImmune
Adam S. Goldstein
Senior Manager, Clinical Purifcation
Operations/Development
Genentech
Brian Lingfeng He
Research Investigator
Bristol-Myers Squibb
Ronald G. Iacocca, Ph.D.
Research Advisor
Eli Lilly and Company
Thomas A. Jennings, Ph.D.
Founding President
Phase Technologies, Inc.
Maik W. Jornitz
Group Vice President
Marketing Fi/FRI
Sartorius-Stedim, Inc.
Hemant N. Joshi, Ph.D., MBA
Principal
Tara Innovations LLC
Daniel R. Klevisha
Director of Pharmaceutical & Chemical Sales
Thermo Fisher Scientifc
Ian Lewis, Ph.D.
Marketing Manager
Kaiser Optical Systems, Inc.
Jack Lysford
Principal Consultant
Lysford Consulting LLC
Steven R. Maple, Ph.D.
Head of Pharmaceutical Technology
Development Dept.
Lilly Research Laboratories,
Eli Lilly and Company
Jerold M. Martin
Senior Vice President, Scientifc Afairs
Pall Life Sciences
John P. Mayer
Senior Research Advisor
Eli Lilly and Company
Mark McDowall, Ph.D.
Strategic Development Manager,
Mass Spectrometry
Waters Corporation
Theodore H. Meltzer, Ph.D.
Capitola Consultancy
Ronald W. Miller, Ph.D., M.B.A.
President , Technology Consultant
Miller Pharmaceutical
Ganapathy Mohan, Ph.D.
Sr. Director, Global Analytical Sciences Dept.
Sanof Aventis
Shane R. Needham, Ph.D.
Laboratory Director
Alturas Analytics, Inc.
Daniel L. Norwood
Director, Physical and Chemical Analysis
Boehringer Ingelheim Pharmaceuticals, Inc.
Yashwant Pathak, Ph.D.
Asst. Dean Academic Afairs
Professor & Chair, Dept. of Pharm. Sciences
College of Pharmacy
Sullivam University
Saeed A. Qureshi
Senior Research Scientist
Health Canada
David Radspinner
Director of Marketing and Applications Support for
BioProcess Production
Thermo Fisher Scientifc
Aniruddha M. Railkar, Ph.D.
Assistant Director
Synerx Pharma LLC
Gary E. Ritchie
Senior Associate
Lachman Consultants
Rodolfo J. Romaach, Ph.D.
Professor of Chemistry
University of Puerto Rico, Mayagez Campus
Jim Rydzak
Investigator, Strategic Technology Division
GlaxoSmithKline
Paul Sheskey
Development Leader in the Water Soluble Polymers
Dow Chemical Company
Charles A. Signorino, Ph.D.
CEO
Emerson Resources
Donald C. Singer
Global Lead Manager, Microbiology, R&D
GlaxoSmithKline
Onkar N. Singh, Ph.D., M.B.A.
Assistant Director, Development Pharmaceutics
Research & Development
Alcon Research Ltd.
Arjen P. Tinke
Principal Scientist, Particle Characterization
Johnson & Johnson Pharmaceutical R&D

Editorial Advisory Board



4 |

|

January/February 2011
124 Bernard E. Saint Jean Drive, East Falmouth, MA 02536 USA
tel: 888.395.2221 fax: 508.540.8680 custservice@acciusa.com www.acciusa.com
Run Both Chromogeni c AND
Turbi di metri c Assays i n
The Same Ki neti c Tube Reader
F L E X I B I L I T Y
CovEr FEAturEs
12 Informatics
The Adaptable Laboratory: A Holistic Informatics Architecture
James M. Roberts, Ph.D., Mark F. Bean, Ph.D., Chris Bizon, Ph.D., John C. Hollerton
& William K. Young, Ph.D., GlaxoSmithKline
22 Powder X-ray Difraction (PXrD)
Approaches to Quantifcation of Amorphous Content in Crystalline Drug
Substance by Powder X-ray Difraction
Peter Varlashkin, Ph.D., GlaxoSmithKline
36 raman
Calibration of Multivariate Predictive Models: The Study of Factors Infuencing the
Prediction Accuracy of Raman Spectroscopy Applied to Pharmaceutical Tablets
Joanny Salvas, Jean-Sbastien Simard, Ryan Gosselin, Ph.D. & Nicolas Abatzoglou, Ph.D., Pfzer
58 single use
Transfer, Implementation and Late Stage Development of an
End-To-End Single-Use Process for Monoclonal Antibody Manufacture
Brian Mullan, Ph.D., Kristi Huntington, Aidan Collins, and Marie Murphy, Ph.D., Eli Lilly & Co.
66 Formulation Development
Amorphous Solid Dispersions as Enabling Formulations for Discovery
and Early Development
Brian E. Padden, Ph.D., Jonathan M. Miller, Ph.D., Timothy Robbins, Ph.D., Philip D. Zocharski,
Leena Prasad, Julie K. Spence & Justin LaFountaine, Abbott Laboratories
January/February 2011 | Volume 14, Issue 1
January/February 2011 |

| 7
30 PArtIClE sIzIng
Physical Characterization of Nano Particulates Used in the
Pharmaceutical Industry
Ron Iacocca, Ph.D.
Eli Lilly & Company
44 DIssolutIon
Limitations of Some Commonly Described Practices in
Drug Dissolution Testing and Suggestions to Address These
Saeed A. Qureshi
Health Canada, Banting Research Centre
50 mICroBIology
Case Studies of Microbial Contamination in Biologic
Product Manufacturing
Kalavati Suvarna, Ph.D, Anastasia Lolas, M.S., Patricia Hughes, Ph.D.
& Richard L Friedman, M.S.
Food and Drug Administration.
74 PrEFIllED syrIngEs/
EXtrACtABlEs & lEAChABlEs
Extractable and Leachable Implications on Biological
Products in Preflled Syringes
FYasser Nashed-Samuel, Dengfeng Liu, Kiyoshi Fujimori,
Lourdes Perez & Hans Lee, Ph.D.
Amgen Inc.
82 EXCIPIEnts
Selling the Audit Observation to Enhance Conformance
Irwin B. Silverstein, Ph.D.
IPEA
sPECIAl FEAturEs
86 Pittcon Advertiser Profles
91 Aguettant Q&A
rEgulAr FEAturEs
90 Industry news
95 Classifed Advertisements
96 Advertisers Index

In thIs IssuE

8 |

|

January/February 2011
SINGLE-USE TECHNOLOGY
Single-use, reusable or hybrid.
The right solution for each process step.
Sartorius Stedim Biotech
USA +1.800.368.7178 | Europe +49.551.308.0
You choose: single-use, reusable or a
combination of both. You set the targets
we provide the technologies to reach
them. Different product types, scale-up
levels and development stages call for
different solutions. Together, we will
simulate your processes and custom-
engineer what best meets your needs.
The result: maximum process reliability,
high fexibility and optimized cost.
www.sartorius-stedim.com/single-use
turning science into solutions
Welcome to the January/February issue of
American Pharmaceutical Review!
I hope 2011 is treating all of you well so far, and that everyone is surviving the winter
storms out there!
I am not one for New Years resolutions, mainly because I give up or give
in by February 1st, so in typical me fashion, this year was no diferent,
no specifc resolution for me, at least not at frst. I vowed to have no
promises to go to the gym every day or to go on a no-carb diet that
will not be fulflled because, quite frankly, I fgure I should always
make sure I exercise regularly and have healthy eating habits, right?
But this year, I did make a diferent kind of expectation for myself. I
made a commitment to myself to make this year a better year than
the ones in the past. Now that may seem vague, and honestly it is,
but my goal is to simply be better, do better, and give more. Why this
pertains to all of you is because this also means that I plan, along with
my colleagues here at Russell Publishing, to make American Pharmaceutical
Review an even better publication than it has been in the past. My hope and
job is to always have the best editorial for our readers, and I will make sure that we
continue to do that tenfold. As always, we are open to your comments and suggestions.
Please let us know how we are doing!
I think we are starting 2011 of right with a great frst round of editorial. James Roberts et al from GlaxoSmithKline are bringing
you their next installment of their two-part series on informatics in the laboratory. In their previous article appearing in our 2010
September/October issue, they supported a laboratory solution that contained both simple and modular applications. Their focus
was on the challenges that arose and what is still needed in analytical laboratories. With this in mind, their second article focuses
on how laboratories can get to where they need to be. Read about their solution on page 12.
On page 58, Brian Mullan et al from Eli Lilly & Co. present an alternate route to biologics manufacturing that creates options for
leveraging existing facilities, as opposed to constructing dedicated biologics manufacturing space. This is a must read for all
Single-Use enthusiasts!
Brian Padden et al from Abbott will discuss their approach for screening and manufacturing amorphous solid dispersions for
application in discovery and early development in their article, Amorphous Solid Dispersions as Enabling Formulations for
Discovery and Early Development. Read all about their work on page 66.
Next up from the University of Sherbrooke is Nicolas Abatzoglou et al on page 36. The engineering goal of their work was to
investigate the development protocol of MVPM-based PAT methods in order to identify the factors that are most infuential on the
performances of the developed method, and thus, to help proposing an optimized development protocol.
And fnally, Peter Varlashkin from GlaxoSmithKline has contributed an article that will briefy mention the various PXRD approaches
to quantify amorphous content, but for the most part, his focus is on a simple approach that does not require standards and is
suitable for routine pharmaceutical development. Check it out on page 22.
Please take a look at the Pittcon Conference and Expo advertiser preview on page 86. I am very much looking forward to seeing
many of you in Atlanta next month!
Kind Regards,
Emily M. Johnson
emily.johnson@russpub.com

"My hope and job is
to always have the best
editorial for our readers,
and I will make sure
that we continue to
do that tenfold"

A message from the Editor



Emily M. Johnson
Editor, American Pharmaceutical Review
10 |

|

January/February 2011
Where do the
key players in pharma,
Biotech and manufacturing
come together?

March 29 - 31, 2011 | jacob k. javits center | new york, new york
INTERPHEXConnects | www.INTERPHEX.com
Interested in exhibiting at INTERPHEX 2011? Contact Pete Zezima at 203.840.5447 or pzezima@reedexpo.com

12 |

|

January/February 2011
InFormAtICs

James M. Roberts
1
, Ph.D., Mark F. Bean
2
, Ph.D., Chris Bizon
3
, Ph.D., John C. Hollerton
4
,
William K. Young
5
, Ph.D.
GlaxoSmithKline, Product Development
1
Research Triangle Park, NC, USA
5
Stevenage, Hertfordshire, UK
GlaxoSmithKline, Molecular Discovery Research
2
Collegeville, PA, USA
4
Stevenage, Hertfordshire, UK
Renaissance Computing Institute, University of North Carolina at Chapel Hill
3
Chapel Hill, NC, USA
The Adaptable Laboratory:
A Holistic Informatics
Architecture
In a previous paper, we advocated a laboratory informatics solution
comprised of simple, modular applications rather than currently
available big box solutions; several examples of efcient and novel
capabilities were provided [1]. Two themes arose. First, limited-
scope, modular applications like those commonly used with personal
computing devices make software easier to use. Second, facile
extensibility of existing software is required to address unanticipated
needs. The content of [1] focused largely on defning what the
challenges of usability and extensibility actually are and what analytical
laboratories need. This paper focuses on how laboratories might
get there, how laboratory software can follow personal computing
capabilities by months, not years or decades. The picture presented is
a holistic one, not relegated to the domain of only a chromatography
data system or only an electronic notebook or only a laboratory
information management system: it is a foundation that can be used
by all of these products as well as for new unforeseen application
solutions. The specifc architecture described is not dogmatic; it
is but one solution, not necessarily the only solution. The principle,
however, is stated axiomatically: analytical laboratories must adapt to
unanticipated needs using data-driven software solutions.
Despite advances in automation and instrumentation, it is inevitable
that customers encounter problems that the vendors have not provided
for. Examples include validation of text input against local business
rules (e.g., project codes, lab notebook references) and vendor-
neutral solutions. Such problems, although related to the analytical
instrumentation, generally lie outside the purview of vendors, yet they
are problems that customers need to resolve. This paper attempts to
suggest an architecture that permits unobstructed, fexible data access,
allowing customers to resolve some of their own problems. It is an
immediate continuation of [1]; the two papers should be read together
and the fgures compared side-by-side. While the discussion has focused
on chromatography and the pharmaceutical industry, the principles and
architecture should be well suited to other market sectors.
Adaptability in Biology and
laboratory Informatics
The laboratory informatics solution described here has similarities to
information fow in biological systems. To illustrate computer concepts
for a broader audience, the architecture is discussed in comparison with
various components in biological systems. While the analogy does not
bolster the proposed architecture, it is hopefully instructive. Like most
analogies, it holds well at a high level but it should not be taken too far.
Biological systems are complex and information rich, far more so than our
laboratory information systems. And yet biology works most of the time.
Organisms are highly optimized for their environment and emergent
properties arise magically from information stored in DNA [2]. Laboratory
P
A
8
3
8
2
1
2
1
0
_
L
New Core-Shell
Phases
REVEALED!


2
0
1
1

P
h
e
n
o
m
e
n
e
x
,

I
n
c
.

A
l
l

r
i
g
h
t
s

r
e
s
e
r
v
e
d
.

K
i
n
e
t
e
x

i
s

a

r
e
g
i
s
t
e
r
e
d

t
r
a
d
e
m
a
r
k

o
f

P
h
e
n
o
m
e
n
e
x
,

I
n
c
.
See Them in Action!
www.phenomenex.com/reveal
Phenomenex products are available worldwide. Email us at international@phenomenex.com.
Ultra-high performance on ANY
LC system just got more powerful.
Introducing two exciting new phases to expand the range
of core-shell selectivity! Kinetex

columns are now available


in fve selectivities: C18, HILIC, PFP, XB-C18, and C8.
XB-C18 C8
PA83821210_L_3.indd 1 1/28/11 10:48:25 AM

14 |

|

January/February 2011
InFormAtICs

informatics systems exhibit similar outcomes; powerful software and


capabilities arise from fles on disk or content stored in memory. Table
1 provides a list of hierarchical components that build on each other to
comprise biological and informatics systems. The similarities lead to one
conclusion: that of an adaptable organism or laboratory.
A holistic Approach to laboratory
Informatics
The overarching theme is that adaptability requires unprecedented
access to the DNA of the laboratory. Throughout this paper, XML is
cited as the document format of choice (the DNA of the lab). This is
not prescriptive; other data-interchange formats like JSON (www.json.
org) or open binary formats might be preferable in some situations.
For brevity, however, XML is cited merely to illustrate the features and
benefts of an open-document based solution. Figure 1 illustrates a
holistic and adaptable computer architecture designed to meet all of
the efciencies and new capabilities described in [1] (e.g., point-and-
click reports, modeling, optimizations) as well as new, unforeseen
applications. At the highest conceptual level, the fow of information
(data and instructions) starts from the bottom left corner of Figure 1.
Upon opening any of the modular client applications, data is validated
before entry into the system and then fows seamlessly through the
entire system in a clockwise direction back to the client application.
In all cases the fow of information is mediated by specialized services
and relational databases.
In summary, data is protected and traceable, satisfying 21 CFR 11
requirements [3]; data is validated before entry into the system
to support unprecedented data mining and downstream error
prevention capabilities and the architecture is rapidly extensible
to support development of new ad hoc solutions or production-
quality applications. Closed, vendor-specifc data formats are not
precluded, but they are also not required because all data is stored
in XML. Finally, data is easily accessible from client applications and,
importantly, from outside these applications using either SQL or
XPath queries. Examples follow, but frst a more detailed description
of the architecture.
A Universe of Possibilities: Te Primary Data Store
The bottom right region of Figure 1, termed the Primary Data Store
(PDS), contains all the laboratory data and instructions, literally
everything that can be known about the system. The XML documents
in the PDS are immutable: once created they can never be changed.
They document the state of every part of the system at a specifc point
in time. New XML documents can be created to supersede existing
ones, but existing documents cannot be modifed or replaced.
The XML documents are produced by the modular application groups
discussed in Figure 3 of [1] and represent sample metadata, analysis
Table 1. Similarities between biological and laboratory informatics
systems in the architecture described here. The analogy is
hopefully instructive for those less familiar with computer systems.
The key point is that both systems can lead to an adaptable
entity that can change to meet the needs of an ever-changing
environment.
Biological
Component
Informatics
Component
Description
DNA XML Contains all the data and instructions,
literally everything that can be
known about the state of the system.
Immutable: portions are copied and
used by other components in the
system, but never modifed.
Gene XML Node A piece of data or instructions. Not all
genes/nodes are used, but they are all
accessible for potential future use by
RNA/services.
DNA Repair Data Validation Corrects mistakes at the source.
Regulatory Genes Business Rules Controls information fow through the
system.
RNA Services Translates data or instructions into a
useful form.
Proteins Relational Databases Specialized components that efciently
carry out a very specifc, useful function
at high speed. Single-purpose, but
interacts with others of its kind.
Cellular Wall Security Protects components from
unauthorized access.
Diferentiated Cell Modular Application Orchestrates activities of a variety of
components to create a specialized,
useful single-purpose function.
Adaptable Organism Adaptable Laboratory The collection of all components
from which emergent properties
arise. Each component can be rapidly
adapted to meet the needs of a
changing environment.
Figure 1. An adaptable architecture supports unanticipated and
changing needs. Every time a modular application is opened
(purple box), data is checked against a Business Rules & Templates
Repository (BRTR). Valid data then fows through modular client
applications into the Primary Data Store (PDS). Specialized
services (orange) respond when new data arrives in the BRTR
or PDS, extracting and placing only the necessary parts into
specialized relational databases that provide organized content
to various client applications. The BRTR and PDS employ a shared,
public ontology; all other components are vendor-specifc. SQL
and XPath interfaces support ad hoc queries. Chromatography
Data Systems, Electronic Notebooks, and Laboratory Information
Systems are all subsets within this architecture.
The new Chromeleon

7.1 Chromatography Data


System brings Operational Simplicity

to the Enterprise.
Fully network all your LC/GC/IC instruments and realize
the benefits of multiple intelligent features. Innovative
eWorkflows enable anyone to run SOPs perfectly with
just a few clicks, and the SmartPeaks

integration
assistant delivers the baselines you want in seconds.
From Samples to Results
Quickly and Easily
Learn more at www.dionex.com/chromeleon7
Chromeleon 7
Simply Intelligent
Chromeleon is a registered trademark, and Operational Simplicity and SmartPeaks are
trademarks of Dionex Corporation. PIN 1042
P
i
t
t
c
o
n

2
0
1
1
R
e
g
i
s
t
e
r

f
o
r

D
i
o
n
e
x

C
o
u
r
s
e
s

C
a
r
b
o
h
y
d
r
a
t
e

A
n
a
l
y
s
i
s

o
r

C
a
p
i
l
l
a
r
y

I
C

w
w
w
.
d
i
o
n
e
x
.
c
o
m
/
p
i
t
t
c
o
n
1
1

16 |

|

January/February 2011
InFormAtICs

metadata, analytical methods, laboratory instrument instructions, raw


instrument data, processed data, and reported data. Binary data (e.g.,
the raw signal) may be encoded in Base64 and stored as a text string
in the XML document or for larger data sets the XML document might
contain a reference to binary data stored in an open, accessible format
to support high-speed access.
Each XML document is uniquely named with a system-generated
human-readable fle name. For example, the XML document
representing processed data might be named as a concatenation of
the unique instrument ID, a timestamp identifying the start of the data
acquisition, and a timestamp representing the start of data processing.
This represents an interpretable and unique fle that can be quickly
found by the services and its data extracted and used by other parts
of the system. Other naming schemes are possible, but they must
ensure a globally unique fle name. As with DNA, not all XML nodes
(genes) are useful for all parts of the system. However, each node is
easily accessible to all other parts of the system. This facile accessibility
is currently missing in todays laboratory and it is the fundamental key
to adaptability discussed in the examples below.
Invisible Helpers: Specialized Services
A service is an invisible software application that runs in the
background. There are two kinds of services (orange box): the minority
that interact directly with a client application (dashed line) and the
majority (solid line) that monitor the creation of new XML documents
on a secure fle share and respond accordingly by safely extracting
useful portions and placing them in a limited-scope relational
database (blue box). This data translation requires a well-defned
object-relational model that is more easily developed if the relational
databases are kept manageably small and of limited scope. Keep it
simple. The services themselves will vary in complexity depending on
scope of the solution, but all of them will likely implement event-based
monitoring and multiple threads to ensure adequate performance
(e.g., flling the database nearly instantaneously upon arrival of
the XML document into the PDS). Note that in addition to simply
shufing data through the system, these individual services can be
licensed as distinct products and combined in numerous ways to form
confgurable workfows using a workfow management application.
Fast Data Access: Specialized Relational Databases
Fueled by the services that provide data from the PDS and BRTR, the
relational database management system (RDBMS) provides fast access
to all the data used by all applications. Two exceptions to this rule
are high-performance applications where a service might provide
content directly to the client from an in-memory cache, bypassing
the RDBMS, or when live streaming data is required (dashed arrow
in Figure 1). Each database stores all superseded records, but by
default applications might expose only the currently valid data from
the PDS and BRTR to prevent cluttering the work environment with
invalid content (e.g., superseded instrument methods). Thus, audit
trails and traceability are available upon special request but old data
does not clutter the current working environment. In principle, a new
database can be easily added, built from the primary data stored in the
immutable XML documents stored in the PDS.
Keeping Data Safe: Security, Authentication, Authorization
Primary data in the XML documents are protected by the computers
operating system. Information stored in the XML documents is
accessible, but only by services and a select few individuals with
appropriate authorization. Modern operating systems provide this
level of security by authenticating (you are who you say you are) and
authorizing (yes, you may have access) user access to content stored
in the XML. Additionally, modern scalable frameworks like iRODS
(Integrated Rule-Oriented Data System), used by the digital archiving,
seismology, astronomy, and physics communities, ofer powerful
security for fle-based collections using granular, policy-based rules
that determine who can do what with a given fle [4].
Poka yoke
getting it right the First time
Todays vendor applications rarely provide sufcient validation of data
entry. Manual, unchecked keyboard entry of text is ubiquitous and
this means that databases are flled with incorrect data or 50 diferent
correct ways to say the same thing; both scenarios result in nearly
useless database queries. And this makes getting meaningful data from
the RDBMS nearly impossible. Data validation is implemented here with
a poka yoke (mistake proofng) mindset, a term borrowed from lean
manufacturing in which errors are ideally corrected before they have a
deleterious efect. The bottom left corner of Figure 1 depicts this feature
implemented using the Business Rules & Templates Repository (BRTR).
Like data in the PDS, these individual XML documents are also immutable,
but updateable with new unique and time-stamped versions to support
traceability. Unlike the PDS, however, the XML documents in this
validation section are singletons: one and only one document instance is
valid at any given time and it is the master that client applications must
consume and conform to every time the application is opened (although
content could be mirrored across multiple computers). It efectively
serves the role of a benevolent dictator, protecting the system from
inadvertent mistakes, because only valid objects are presented to the
users client for downstream entry into the PDS and relational databases.
Structurally, this could be implemented as a dictionary containing key-
value pairs. The values can be simple or complex depending on how
many variables are needed to describe the object. For example, the
values for an HPLC column might contain one and only one validated
value for each of the following: vendor, model, part number (not serial
number), length, diameter, particle size, stationary phase, and minimum
and maximum values for pH and temperature. Validated values for
these objects leads to extremely positive downstream afects: fast
meaningful aggregation queries, intelligent error checking in the lab,
instantaneous global adoption of business rules, and point-and-click
report generation. Four control mechanisms exist in the BRTR and are
summarized in Table 2.
Job #: WATR13531_A
Job Name: WATR13531_A_Bioanalysis_DBmec_edit.indd Date: 01-26-11
Live: 6.75"x9.5" SmallTrim: 7.75"x10.5 LargeTrim: 8.375"x10.875" Bleed: 8.625"x11.375" Page: 1 Rev: 1
Stage: mech Release: 01-26-11 DB AD: CP CW: GK PM: JT Scale: 100%
Color: 4C Gutter: Other: ST: DB/JC
MK: _____________ PM: ______________ AD: ______________
CW: _____________ QA: ______________ CD: ______________
Initials / Date Initials / Date Initials / Date
Client / Date:
________________________________
MECH
PROOF
A
P
P
R
O
V
A
L
S
PUBS:
American Laboratory
American Drug Discovery
American Pharmaceutical Review
Bio Business
Bio IT World
BioTech World
BioTechnology Focus
BioPharm International
BioProcess International
BioSpectrum Asia
Chemical & Engineering News
Drug Discovery and Dev
Drug Discovery News
Environmental Science & Technology
Food Processing
Food Quality
Food Safety Magazine
Genetic Engineering News (GEN)
Genome Technology
Genomics & Proteomics
Jour of The American Soc for
Mass Spectrometry
Lab Asia Media Guide
Lab Business - Jesmar
LCGC Asia
LCGC N. America
LCGC Europe
Molecular and Cellular Protemomics
Nature Methods
Pharmaceutical Discovery
& Development
Pharmaceutical Manufacturing
Pharmaceutical Technology
Pharmaceutical Technology EUROPE
Pharmaceutical Executive
Pharm Form and Quality
Proteomics Journal
Scientific Computing & Instrumentation
Scientific Computing World
Please note this is a COMMON SIZE mechanical file,
you will need to center file using the center marks
provided when placing ad in the publication page area.
(common size = smallest live/smallest trim/ largest bleed)
2011 Waters Corporation. Waters is a registered trademark of Waters Corporation.
The Science of Whats Possible is a trademark of Waters Corporation.
Have you seen the worlds most comprehensive Bioanalysis system solution?
Waters has introduced the technology to tackle any challenge, as well as
transform your laboratory. And now were taking it on tour.

18 |

|

January/February 2011
InFormAtICs

Adaptability in Action: Examples


from the real World
Instrument Control & Data Acquisition
Real-time, closed-loop automated control of laboratory instruments
was described in [1]. These and other intelligent applications require
a new level of instrument control. Figure 2 shows how this architecture
can support rapid development of new experiments in the lab, where
the instrument is treated as a black box that receives instructions from
Application Group 2 [1] in the form of an XML document containing a
sequence of instructions.
In this model, the primary job of the laboratory instrument is to execute
instructions provided by the XML document, raise events to notify
clients that something important has happened, and raise exceptions
if something goes wrong so clients may handle errors appropriately.
For example, if the instrument is an NMR, the instruction set might
be sample metadata along with a series of shaped RF pulses, delays,
and acquisitions combined to make a novel pulse sequence; for
HPLC, one single instruction could be turn on column heater and
heat to 40 degrees C. All of these instructions are mediated by the
instrument-vendors instrument control service that reads the simple
XML instructions and implements a max-priority queue [5] of sample
sequences, bundled with the instrument instructions associated with
each sample in the sequence. All the complexity of instrument control is
hidden and handled by the service. The idiom is, Heres the sample and
its method information, go! This approach amounts to a facade design
pattern [6, 7] that simplifes todays proprietary instrument drivers and
is akin to a specifcation-implementation model used for computer
language compilers. Diferent analytical techniques will obviously
require diferent instruction sets and vendor-specifc hardware will
require slight diferences in instruction sets based on diferent hardware
features or capabilities. However, all instructions should be defned at
a level of abstraction to support adaptable work practices by analytical
chemists, not software developers.
Data acquisition is handled by the instrument vendors service and has
two distinct outputs. First, during data acquisition a typed (decorated)
binary stream is accessible to enable reading of temporary, non-
stored live data as it is generated from the instrument. Second,
upon completion of data acquisition, the raw data produced by the
instrument is written to the PDS in Schema Group 3 (see Figure 1).
This level of instrument control and data acquisition allows for
intelligent, live, interactive method development, optimization, and
error correction on-the-fy, without the need for manual intervention.
This automated closed loop of control instrument analyze data
control instrument ofers unprecedented opportunity to automate
extremely time-consuming manual activities (e.g., optimization) and
it is easily supported using this approach.
Data Access Concept
Two sources exist for ad hoc data access: the relational databases
and the XML documents. This two-option data access model exploits
the strengths and avoids the weaknesses of both XML and RDBMS
as data storage mechanisms. For example, XML encapsulates
Table 2. Business rules are enforced globally and immediately
before data enters the system because each client application
checks for updates to confgurations, business rules, and templates
every time the application is opened for use. This poka yoke
error prevention and correction enables instant adoption of
changes in business rules and SOPs across an entire organization.
Control Mechanism Purpose Examples
Application Confguration
& Installation Packages
Ensure that the correct
version of software is
used and role-based
permissions are enforced.
application version, user roles
and authorization, granular
permissions, installation packages
to update applications
Content Validation Ensure that business rules
are applied uniformly
and immediately.
analytical techniques, analytical
methods, instrument identifers,
sites, buildings, laboratories,
addresses, people, teams, date and
time formats, projects, columns,
packaging materials, storage
conditions, reagents, solvents,
batches, chemical structures,
calculated chemical properties,
physical constants
Analysis Template Ensure that all
experiments are carried
out according to
appropriate scientifc and
business standards.
purpose-conclusion, method
development, method validation,
dissolution, content uniformity,
excipient compatibility, solubility,
stability, reaction monitoring,
identity, purity, assay, structural
elucidation, method transfers,
robustness, ruggedness,
disintegration, friability, particle
size distribution, heavy metals, loss
on drying, residual solvents, ash,
water content
Report Templates Ensure that dynamically
generated point-and-
click reports conform
to currently accepted
internal business rules or
external guidance.
certifcates of analysis, batch
impurities, stability, column
logging, instrument utilization
metrics, IND, NDA, IMPD, MAA
Figure 2. Specifcation-implementation model for laboratory
instrument control and data acquisition. Instrument vendors
implement instructions provided to the instrument control
service by Application Group 2. Data is saved to the PDS as raw,
unprocessed data for consumption by Application Group 4 (Data
Processing, see [1] and Figure 1 above). Live data streaming
from the instrument is provided as a decorated stream for client
applications to display and respond to live data.
A Clear Road Ahead for Metabolite ID
Complimentary technology for your evolving drug metabolism needs.
Whether your needs revolve around low-level metabolite identifcation or high resolution accurate mass structural elucidation,
the AB SCIEX QTRAP

5500 and the TripleTOF

5600 mass spectrometry systems can make your drug metabolism goals a reality.
See how clear the road ahead for metabolite ID can be with targeted predictive MRM and non-targeted full scan MIMs approaches.
Discover how HRMS Qual-Quant workfows can accelerate and simplify complete metabolite characterization and metabolic
clearance in a single analysis.
For more information, visit www.absciex.com/applications/drugmet
For Research Use Only. Not for use in diagnostic procedures.
2010 AB SCIEX. The trademarks mentioned herein are the property of AB Sciex Pte. Ltd. or their respective owners. AB SCIEX is being used under license.
AB SCIEX TripleTOF

5600 System AB SCIEX QTRAP

5500 System

20 |

|

January/February 2011
InFormAtICs

everything there is to know about a single object one sample, one


sample sequence, one experiment, one instrument, etc. in a single
document. This gives rise to its primary strength all the data that
describes the object is accessible from one neat package and XML can
be intuitively structured to access anything and everything from the
single XML document. XML is also descriptive; it conveys meaning and
is easily interpreted. So if you want to know everything about a few
objects, simply use one of many available third-party tools to query
the XML. However, XML is not ideal for working with extremely large
collections of similar objects. Enter RDBMS, which organizes data in
tables and provides superior data access to large collections of similar
objects, spread across many diferent tables. The primary beneft of
this organization is the ability to execute arbitrary and unforeseen
queries across multiple objects in an efcient manner. The weakness
of RDBMS, however, is that without clear documentation describing
the database, it can be difcult or impossible for a non-specialist to
interpret, and therefore very difcult to actually create new queries.
While the greater generality and efciency associated with multi-
object queries often tips the tables towards exclusive use of RDBMS,
domains in which data needs are well understood, and which are
primarily single-object based (as with many laboratory activities) can
beneft from the usability of the document format. Furthermore, a
document model ofers a convenient way to aggregate data, e.g., from
multiple vendors. If vendors each create a separate database, it can be
technically challenging to query across these databases; a document-
based aggregation, though it may be somewhat inefcient, is trivially
achieved. In the examples that follow, this pattern of moving between
RDBMS and XML data formats emerges as an extremely fexible
solution to the adaptability challenge facing todays laboratories.
Data Access Example
Its a common scenario. A manager or scientist asks a question that
requires data spread across multiple systems or locations. Figure
3 illustrates a rapid workfow that can provide fast answers to
unanticipated questions, based on using the SQL and XPath interfaces
specifed in Figure 1.
If the necessary data exists in the RDBMS, then an ad hoc query is
executed and an ad hoc result set is produced to satisfy a one-of
need, usually in a few minutes. Custom queries will proliferate and
over time these read-only SQL queries will form a collection that
can be monitored for frequency of use or relative importance. If
necessary, a stored procedure is easily added to the RDBMS, thus
enabling a generalized answer that can be executed in seconds.
The second possibility is that the required data is not stored in the
database (the No path in Figure 3). In this case, the PDS provides the
data. Individual XML documents on the fle share can be iterated over
by operating system services and needed information contained in each
XML document can be extracted using an XPath query. This pattern
supports both DOM and SAX models, optimized for either full-control
or high-speed data access. The end-result is the same as if the data were
stored in the RDBMS: an ad hoc result set is provided quickly. As with the
RDBMS data access example, frequently requested XPath queries can
be prioritized and implemented as a new data service, relational data
table(s), or even a new database, each of which would be optimized
to support the custom (but important) data access needs. To continue
the biological analogy, introns (previously unused XML nodes) have
become exons (useful XML nodes).
Point-and-Click Reports
Writing reports and documents manually is extremely time-
consuming. This architecture supports the rapid development of new
reports that can be generated with a few mouse clicks. For example, an
analyst responsible for creating a 6-month stability report for a batch
of tablets might simply start to enter the tablet batch number in the
reporting application in Application Group 5, which auto-completes
the text entry midstream (because the values are validated). The
analyst then chooses the 6-month time point from a drop down list
and clicks a button to request the report, which is generated and
provided in a few seconds (the data being aggregated on-the-fy
by the database). Importantly, all the content of the report is valid,
enforced by the current Report Template. Likewise, the presentation
(table formats, signifcant fgures, charts, fonts, headers, footers, etc.)
also conforms to currently accepted criteria, enforced by the Report
Template. In short, an arbitrarily complex report can be accurately
created in a few seconds. Thus, the analyst stops spending countless
hours fnding data, transcribing data, formatting data, building charts,
and creating reports manually. Likewise, managers stop tedious
review of data transcription. Instead, time is spent understanding the
meaning of the data, identifying trends or outliers, and taking action
based on the impact the data has on the project.
Figure 3. Extensible data access using both relational databases
and XML documents in the PDS. Unanticipated questions can be
quickly answered using a SQL query against a relational database.
Subsequent ad hoc answers then accumulate over time; those that
rise in importance can be implemented as a stored procedure for
fast data access. If the data does not exist in the database, the PDS
is queried after which important queries then get supported by a
new database.

January/February 2011 |

| 21

InFormAtICs
What We need:
getting from here to Tere
In this two-paper series, we have tried to be explicitly clear about
new, untapped opportunities that await the laboratory. We have
also described a potential mechanism (not the only one) to make
these concepts a reality. The architecture proposed here is diferent
from many of todays laboratory software oferings. It is a more open
approach to accessing data and controlling laboratory instruments,
but it is also more modular with opportunities for new commercially
licensed products. In this closing section, the main challenges and
next steps toward adoption are summarized.
Open, Shared Schema
The architecture proposed here requires a shared language, a
foundation on which everything else rests: the content stored in the
document-based PDS and BRTR. This amounts to a holistic, open
and shared schema a common defnition used by all instrument
vendors, similar in spirit to the open, vendor-neutral AnIML instrument
data format [8]. Successful implementation and adoption of these
standards will require iterative development and an unprecedented
level of collaboration between leaders in industry, academia,
instrument vendors, and regulatory agencies.
Closed, Proprietary Implementation
Everything above the BRTR and PDS in Figure 1 can, and should be, a
vendor-specifc, commercially licensed product. Competition at the
level of services, databases, and modular applications will produce
the best products for customers. Using the shared schema, software
vendors are free to diferentiate themselves and compete for customers
at this new level: that of the modular application and service. From
a vendors perspective, the smaller, modular applications can be
confgured and combined in nearly limitless ways to gain customers
in new markets: CROs, small biotech and large pharmaceutical
companies, academic and government labs, and diferent chemical
industries can all have variations, using the same foundation.
Next Steps:
An Open Discussion of Risks, Costs and Benefts
From an instrument software vendors perspective, there are several
risks and benefts associated with adopting this approach: loss of
closed data formats, loss of vendor-specifc instrument control,
its been tried before. We do believe, however, that there are valid
fnancial incentives for moving in this direction, not the least of which
is that someone will do it (consider the MP3 fle standard and the
music industry). These risk-beneft-cost issues are too complex to
address in a short communication and the authors believe that these
topics are best discussed in person. What is needed is a mechanism
to bring together representatives from industry, academia, instrument
vendors, and regulatory agencies to discuss how laboratories can
move toward more open, scalable and adaptable work practices.
Please contact us if you have questions or criticisms.
references
1. Roberts, J.M., Bean, M.F., Cole, S.R., Young, W.K., Weston, H.E., American Pharmaceutical
Review, September/October 2010, p. 60 67.
2. Complexity: A Guided Tour, 2009, Oxford University Press, Oxford, Melanie Mitchell.
3. Code of Federal Regulations Title 21, Part 11.
4. Proceedings, iRODS User Group Meeting 2010: Policy-Based Data Management, Sharing and
Preservation, Edited by Reagan W. Moore, Arcot Rajasekar, Richard Marciano.
5. Introduction to Algorithms, 2nd Edition, 2001, The MIT Press, Cambridge, MA, p 138 140,
Thomas H. Cormen, Charles E. Leiserson, Ronald L. Rivest, Cliford Stein.
6. Head First Design Patterns, 2004, OReilly Media, Inc., Sebastapol, CA, p 254 270, Eric
Freeman, Elisabeth Freeman.
7. Design Patterns: Elements of Reusable Object-Oriented Software, 1995, Addison-Wesley,
Reading, MA, p 185 193, Erich Gamma, Richard Helm, Ralph Johnson, John Vlissides.
8. See http://www.scientifccomputing.com/the-iupac-astm-unifed-standard.aspx.
Author Biographies
Dr. James M. Roberts works in an automation group in Product
Development at GlaxoSmithKline. He applies informatics, data
integration, modeling, and automation to increase efciency in analytical
chemistry. He received a Ph.D. under the advisement of Professor Janet
Osteryoung and has 12 years of experience in the pharmaceutical industry.
Dr. Mark F. Bean, Investigator at GlaxoSmithKline, has worked with
automated MS and software solutions for research chemistry LCMS for 20
years. He was the architect-project lead for CANDI, a vendor-neutral LCMS
login, processing, and results viewing suite used by GSK in the Philadelphia
area. He is a founding member of the ASTM committee charged with the
AnIML analytical data standard.
Dr. William K. Young is an Investigator in Analytical Sciences at
GlaxoSmithKline in Stevenage, UK. He develops and maintains the walk-
up chromatography systems within Chemical Development. He received
his Ph.D. from Imperial College, London with Professor W. John Albery and
has 11 years experience in the pharmaceutical industry.
John C. Hollerton is a Director of Analytical Chemistry at
GlaxoSmithKline. He leads a team who offer spectroscopic and
small molecule X-ray crystallography support to R&D as well as
informatics support to Analytical Chemistry. He has spent the last
30 years integrating informatics in the analytical environment. He
was responsible for the design and implementation of GSKs Global
Analytical Data Repository (GADR).
Dr. Chris Bizon is a Senior Research Scientist in Informatics at the
Renaissance Computing Institute. He currently leads a group designing
cyberinfrastructure for high-throughput genomic sequencing. He received
his Ph.D. in physics at the University of Texas at Austin with Professor Jack
Swift and has worked in both the academic and industrial settings

22 |

|

January/February 2011
PXrD

Peter Varlashkin, Ph.D.


GlaxoSmithKline, US R&D
Approaches to Quantifcation
of Amorphous Content in
Crystalline Drug Substance by
Powder X-ray Difraction
Introduction
Typically, the desired solid state for active pharmaceutical ingredients
(drug substance) is crystalline. Amorphous content within the crystalline
drug substance may have a deleterious impact on the drug product such
as reduced chemical and physical stability.
Powder X-ray difraction (PXRD) is one of several signifcant tools
used to characterize the solid state character of drug substances.
Randall et al [1] discussed the various uses of PXRD to pharmaceutical
development as applied both to drug substance and drug product.
Powder X-ray difraction is considered the gold standard for identifying
and quantifying crystalline phases in materials as diverse as minerals to
active pharmaceutical ingredients. For pharmaceutical samples with
amorphous content within the range of approximately 10 to 90% w/w,
powder X-ray difraction (PXRD) provides a means to determine the
relative amorphous content. While less-routine XRD techniques may
provide a lower limit of detection/quantitation for amorphous content,
in the authors experience, 10% amorphous content is a practical lower
limit for detection using typical PXRD equipment.
The purpose of this article is to briefy mention the various PXRD
approaches to quantify amorphous content. The majority of this
article will focus on a simple approach that does not require standards
and is suitable for routine pharmaceutical development.
Experimental
The data presented were obtained with a typical Bragg-Brentano
powder difractometer using a copper X-ray source and a nickel flter at
the detector to provide difraction data using = 1.54 .
Discussion
During the development of a drug substance synthesis, initial material
may be completely amorphous or poorly crystalline. PXRD can be used
to screen for the relative crystallinity of samples. At some point, it may
be advantageous to determine quantitatively the level of amorphous
content in the samples.

January/February 2011 |

| 23

PXrD
Quantitative phase analysis by PXRD is not trivial [2]. However, the
complexities of analyzing a material such as a geologic sample are
lessened for organic samples such as pure drug substance or drug
product due to minimal sample X-ray absorption afects.
Figure 1 shows the PXRD pattern for a mixture of crystalline and
amorphous drug substance X. In a mixture of amorphous and
crystalline material, the PXRD will exhibit both sharp and broad
features. The sharp peaks are due to the crystalline component and
the broad features (sometimes referred to as halo) to the amorphous
component. Deconvolution of the mixture PXRD into separate PXRDs
with sharp-only difraction peaks and broad-only difraction peaks will
allow for determining the percentage of amorphous content.
Prior to applying quantitative methods to PXRD data for amorphous
determination, the analyst must be sure that the broad features
are due to amorphous (or glassy) material rather than due to
microcrystalline or nanocrystalline material. The difraction peak
shape is afected by a variety of parameters, including the size of
crystallites making up drug substance particles. As the crystallite
size is reduced, the difraction peaks broaden. Once the size is
sufciently reduced, the crystalline difraction peaks may have
broadened to the extent to merge into each other forming a single
broad difraction peak (halo). If a single-crystal X-ray difraction data
is available, the powder difraction pattern can be calculated with
diferent peak shapes and widths to simulate the PXRD of micro/
Figure 1: PXRD scan of sample X obtained using a back-flled cavity
mount and a variable divergent slit. Scan collected in refection
mode. The sharp features (peaks) are due to the crystalline
component. The broad hump in the baseline is due to the
amorphous component. Background scattering unrelated to the
amorphous and crystalline components produces an ofset of the
baseline from 0 counts.
X-ray diffraction solutions for
the pharmaceutical industry
EMPYREAN
Widest range of pharmaceutical applications:
Structural investigation of NCEs
Polymorph and salt screening
In situ crystallization and scale-up studies
Investigation of amorphous and nano-crystalline
compounds
Stability and compatibility studies
Detection of low amounts of polymorphic
impurities
Determination of small to large amorphous content
Microstructure analysis of tablets by CT
For more information:
info@panalytical.com
www.panalytical.com
The Analytical X-ray Company
Microstructure analysis
of counterfeit and
original tablet
10
0
2500
10000
22500
I
n
t
e
n
s
i
t
y

[
c
o
u
n
t
s
]
2 Theta []
20 30
10%
30%
50%
90%
XRD scans
for samples
with different
amorphous
lactose content
V
is
it u
s
a
t P
ittc
o
n
b
o
o
th
2
2
6
1
PN7722.indd 1 04-02-2011 07:57:14

24 |

|

January/February 2011
PXrD

nanocrystalline material. Such material would be crystalline but


less ordered due to the reduced repetition of molecular packing in
3-dimensional space compared to material exhibiting PXRD with
sharp difraction peaks. Analysis for microcrystalline material is a
separate topic not discussed here. In the absence of single-crystal
XRD data, the analyst should discuss with the sample submitter the
history of the sample to understand if assaying for true amorphous
content is appropriate. The presence of a glass transition in the
diferential scanning calorimetry profle of the material would serve
as proof of the presence of amorphous material.
At the early stage and even later stages of development, it would
be advantageous to use a method for determining amorphous
content that did not require the use of standards. This approach
is discussed next.
Approach 1 PXRD Quantitation of Amorphous
without the Use of Standards
For samples where little material (tens of mg) is available, silicon or
quartz zero background plates may be used. Typically samples are
dusted on the plates previously coated with a thin flm of petroleum
jelly or other viscous material. The level of the petroleum jelly for
PXRD sample preparation is typically kept low to minimize the signal
from the petroleum jelly which might be confused for amorphous
drug substance. For the analysis of amorphous content, the use of
petroleum jelly should be avoided. Thus, the sample should remain
horizontal during the difraction scan which is a feature of theta/theta
difractometers. Theta/two-theta difractometers require the sample to
tilt with the scan which may cause the powder to fall of the stage. Sample
spinning to improve counting statistics makes the problem worse so
the use of theta/theta systems for zero background plate preparations
is recommended.
Where more material is available (200 mg or greater), the sample
material may be flled into a sample cavity. For quantitation of
mixtures of crystalline phases, back-flling [3] is recommended to
avoid the efects of preferred orientation of particles when the sample
is flled from the top of the cavity and smoothed fat with a glass
slide. For the quantitation of amorphous/crystalline mixtures using
the complete difraction pattern, preferred orientation issues are less
important. Thus front-flling of the sample is an option if back-flling
is not available or problematic. A comparison of the reproducibility of
various sample preparations on the raw and processed difraction data
is recommended.
Modern difractometers allow for the data collection to be obtained
with either fxed or variable divergent slits. The slit situated in front
of the X-ray source, limit the divergence of the X-ray beam. PXRD
results collected using a fxed divergent slit is also known as constant-
volume data. Starting from low two-theta and scanning to higher
two-theta, the area illuminated by the X-ray beam gets smaller and
the depth of penetration increases. Provided the area illuminated
with X-rays does not exceed the sample boundaries and the sample
depth is sufciently thick that the X-ray beam will not penetrate to the
bottom of the sample at the highest scanned difraction angle, the
sample volume interacting with the X-ray beam remains constant.
PXRD results collected using a variable divergent slit is also known
as constant-area data. The divergent slit is decreased as the scan
goes to higher difraction angles which results in a constant area of
sample illumination but diferent sample penetration by the X-rays
with varying difraction angle. For a thin zero background plate
preparation, the X-ray beam will completely penetrate the sample
such that both a constant sample area and a constant sample volume
are analyzed.
For cavity-amounted samples, the fxed divergent slit operation may
be preferred so that a constant volume is analyzed throughout the
difraction scan. For zero background plate preparations, the variable
divergent slit approach is recommended. In practice, data can be
collected both ways during method development.
Where sufcient sample is available, a cavity-flled sample preparation
is recommended to maximize the amorphous signal relative to the
background.
In addition to the typical refection geometry, modern research grade
difractometers allow for the option of collecting the difraction data in
a transmission mode. This alternate mode may be considered during
method development.
For routine PXRD, the typical difraction scan range is around
approximately 2 to 45 degrees two-theta (with copper K-alpha
radiation). Most PXRD vendor software allows the user to deconvolute
the PXRD pattern to isolate the crystalline component. A blank
scan should be run to determine the background artifacts from the
difractometer. Depending on the difractometer optics, the lower
difraction limit for integration of the difraction signal from the
amorphous and crystalline components will be approximately 3 to 4
degrees two-theta (to avoid scatter from the direct X-ray beam at low
angles). The upper scan limit should be approximately 70 degrees to
allow proper estimate of the PXRD baseline.
Figure 2 shows the resulting deconvolution of the sample PXRD scan
presented in Figure 1. Diferent vendor software allow the user to
adjust the resolution to discriminate between the gradual baseline
changes due to amorphous content and background scatter. By
subtracting out the portion of the data above the baseline that
separates the crystalline from amorphous components, the difraction
intensity for the amorphous component can be determined. The ratio
of the difraction intensity for the amorphous component divided
by the total difraction intensity multiplied by 100 provides the
approximation of the percent weight/weight amorphous content. The
starting point (low two-theta) for integration is just above the point
where scattering from the direct beam is observed. The ending point
is where the difraction signal returns to the background level.
The approach may result in background signal due to X-ray scattering
by air and the sample holder being incorporated into the difraction
area associated with the amorphous component. The deconvolution
process can be refned by running blank scans. For samples prepared
using zero background plates, the blank scan would simply use a clean
zero background plate with no applied sample. For samples prepared
using cavity mounted sample holders, an empty sample holder is not
appropriate since scattering from inside the sample holder may be
Innovation with Integrity
Analytical Power and Speed for:
Small Molecule Identication
Trace Component Analysis
Intact Protein Analysis
Peptide Identication and Sequencing
Characterization of Post Translational Modications
X-RAY POWER for PHARMA:
Phase/polymorph identication and quantication
Non-ambient studies including computer-controlled humidity
Structure solution and analysis from powder data using TOPAS
High-throughput solutions from the XRD market leader
maXis D8 Advance
LC-MS / XRD
www.bruker.com/d8advance
www.bruker.com/ms
C
M
Y
CM
MY
CY
CMY
K
APR Bruker Dual Ad Full Page print.pdf 2/4/2011 4:28:42 PM

26 |

|

January/February 2011
PXrD

observed. If available, a zero background plate inserted into the cavity


can serve as a blank. The height of zero background plate should be
fush with the top edge of the cavity. Applying a small piece of modeling
clay afxed to the underneath side of the plate and then turning the
sample holder over and pressing against a smooth and clean surface
(such as as a fat bench top or a glass slide) can help properly align the
plate with the cavity top. Inspection of the blank scans would allow
for estimating the instrumental background and then subtracting this
from sample scan prior to deconvolution of the sample difraction
pattern into the crystalline and amorphous components. The PXRD
scan presented in Figure 2 had the instrumental background removed
prior to deconvolution into amorphous and crystalline components.
The lower the percent amorphous, the more attention to detail in
background removal and integration parameters is required.
If possible, a sample containing a sizeable fraction of amorphous
content (30 to 70%) should be analyzed by 13C solid-state nuclear
magnetic resonance (SSNMR). SSNMR is a nuclei-counting technique
and does not require external standards to quantitate amorphous
content. The results from the SSNMR can be used to check the PXRD
results and aid in optimizing the PXRD processing parameters, the
choice of sample preparation, the choice of variable or fxed divergent
slits, and the choice of refection or transmission data collection. The
SSNMR results are most trustworthy when there is a clear separation
in the resonance amorphous and crystalline peaks. If there is overlap
requiring deconvolution to separate the amorphous and crystalline
components, then there is added uncertainty in the percent
amorphous calculation. If the overlap in the SSNMR data is severe,
then SSNMR may not be useful in providing an orthogonal check of
the PXRD method.
Vibrational spectroscopy (IR and Raman) can also be used to check the
PXRD method. However, the IR and Raman methods require external
calibration curves that would involve preparing mixtures of pure and
amorphous standards. Lack of homogeneous mixing, particle size/
morphology diferences between the calibration curves and actual
samples, and diferences in the nature of the amorphous standard and
amorphous content in the sample will lead to errors in the estimate of
the percent amorphous.
Preparation of known w/w percent mixtures of amorphous and
crystalline material can also be used to check the PXRD method
with the same caveats as presented for analysis using vibrational
spectroscopy. For example, an amorphous standard prepared by ball-
milling, spray drying, or lyophilization may not be fully equivalent to
actual amorphous content produced in the sample by some other
process (e.g. micronization using an air-jet mill or fast precipitation of
the drug substance during crystallization).
Approach 2 PXRD Quantitation of Amorphous
with External Standards
As mentioned before, mixtures of amorphous and crystalline materials
may be prepared. If a pure amorphous standard is not possible, a
partially amorphous standard (e.g. 60%) may be used but would
require estimation of the percent amorphous content by PXRD using
Approach 1 or an orthogonal technique such as SSNMR. The same
approach can be used if a pure crystalline standard is unavailable.
Each mixture can be prepared and then analyzed by method
in Approach 1. The calibration curve would plot the percent
amorphous, as determined by Approach 1 on the y-axis versus the
weight/weight percent from the known weights of amorphous and
crystalline material used to prepare the calibration mixtures. The
calibration curve should be approximately linear allowing for a least
squares fit. From the calibration curve, the observed % amorphous
based on the area responses of the crystalline and amorphous
components can be converted into a weight/weight percentage.
A simplifed variation on Approach 2 is to use the peak intensity of a
single XRD peak for the crystalline component and the intensity at a
two-theta position where signal from amorphous content is present
but there is not an overlapping crystalline XRD peak. The single XRD
peak should be at a low enough two-theta value where signifcant
signal from the amorphous is absent or the baseline intensity near
the XRD peak can be used to subtract the amorphous signal from the
XRD (crystalline) peak intensity. Preferred orientation and size of the
difraction domains of the crystalline component need to controlled
[3]. With XRD deconvolution tools available, use of the full difraction
pattern is preferred.
With all the caveats using mixtures of external standards, the approach
without standards is recommended. Many analytical results (e.g. %
area-under-the curve by HPLC, particle size distribution) produce
results that are relative and not absolute but still allow for batch-to-
batch comparison and data trending.
Figure 2: PXRD scan of sample X following deconvolution of the
difraction pattern into the crystalline (portion above green trace)
and amorphous (portion below green trace) components.

January/February 2011 |

| 27

PXrD
Approach 3 Use of Rietveld Analysis for
Quantitation of Amorphous
If the single crystal X-ray difraction structure is available, Rietveld-
based methods that use the whole difraction pattern may be
employed to estimate the percentage of amorphous content in drug
substance [2]. The approach would require spiking the sample with an
internal standard where the crystal structure is known such as silicon
powder. The data should be collected using a fxed divergent slit. The
analysis involves calculating the difraction patterns for the crystalline
drug substance and the internal standard and then varying the
relative amounts of each component until good agreement between
the observed and calculated difraction patterns for the spiked sample
are obtained. The use of the internal standard allows the Rietveld
analysis to provide the weight fraction of the crystalline component.
Subtraction of this value from 1 would yield the estimated weight
fraction of the amorphous component. Approach 3 can also be an
alternate approach to check the results from Approach 1 or 2. The
use of Rietveld approach for quantitation of amorphous should not
be undertaken by the novice XRD user and thus no further details are
provided. For the more sophisticated user, Rietveld tools are typically
part of the software of modern research grade difraction systems.
For samples with amorphous content below 10%, the difculty in
obtaining reliable results by PXRD increases and alternate techniques
to assess amorphous content, such as gravimetric vapor sorption or
thermal analysis, are suggested. For samples with crystalline material
below 10%, Approaches 1 or 2 may be sufcient. For higher precision,
standard addition methods by spiking the sample with crystalline
material may be required.
Conclusions
PXRD represents a convenient method to determine amorphous
content over a broad range. The use of the approach without
standards is convenient and often sufciently accurate to help drive
the optimization of the drug substance synthesis.
Acknowledgement
The author thanks Graham Whitesell of GSK for reviewing
this manuscript.
Enhance Productivity
PDF-4/Organics 2011
A comprehensive materials database featuring
~436,000 organic and organometallic phases.
Designed for rapid materials identification
Polymorph Screening
Quality Control
Drug & Excipients Identification
Formulation Analysis
Quantitative Analysis
Polymorph Identification
Crystallite Size
XRPD, a nondestructive
technique, coupled with our
comprehensive database is
useful for preformulation,
drug development, and
patent applications.
International Centre
for
Diffraction Data
Databases from the
Database Experts
www.icdd.com
marketing@icdd.com
Phone: 610.325.9814
Toll-free: 866.378.9331 (U.S. & Canada)

28 |

|

January/February 2011
PXrD

references
1. Randall, C., Rocco, W., Ricou, P., XRD in Pharmaceutical Analysis: A Versatile Tool for
Problem-Solving
2. Powder Difraction, Theory and Practice, Dinnebier, Billinge, S. (eds.), Chapter 11
(Quantitative Phase Analysis, authors Madsen, I, Scarlett, N.), The Royal Society of Chemistry,
RSC Publishing (2008)
3. A Practical Guide for the Preparation of Specimens for X-ray Fluorescence and X-ray Difraction
Analysis, Buhrke, V., Jenkins, R., Smith D. (eds), Wiley, VCH, 1998.
Biography
Peter Varlashkin received his PhD in Analytical Chemistry from
the University of Tennessee (Knoxville). He has been employed by
GlaxoSmithKline (GSK) for over 22 years. He currently works in the
Physical Properties group within GSK (RTP, NC, USA). He has published
several papers on powder X-ray difraction and is on the organizing
committee of the Pharmaceutical Powder X-ray Difraction Symposia as
well as a member of the International Centre for Difraction Data.
Become an author.
American Pharmaceutical Review strives to inform readers of the latest information in the
pharmaceutical industry. If you are employed at a major pharmaceutical company or university, or
are an established consultant in the pharmaceutical industry, you are exactly who we are looking for.
All articles submitted must be unbiased, not mentioning specifc vendor companies or
product names, and of a technical nature. For more information on becoming one of
our authors, contact us at 317-816-8787 or emily.johnson@russpub.com.
www.americanpharmaceuticalreview.com
Untitled-1 1 1/21/11 6:33:24 PM

30 |

|

January/February 2011
PArtIClE sIzIng

Ron Iacocca, Ph.D.


Eli Lilly & Company
Physical Characterization
of Nano Particulates Used
in the Pharmaceutical
Industry
Abstract
Nano particulates or nano-scaled pharmaceutical products have
received great interest for the past two decades. Reduction in size to
the nano scale can ofer unique therapeutic advantages, such as the
ability of particles to pass through cell membranes, thereby targeting
specifc cells. There has also been a great deal of research performed
on the use of nano particles for imaging specifc tissues or tumors
in vivo. The physical characterization of these nano materials poses
certain challenges to the scientist/engineer working in this feld. This
article highlights these analytical challenges and proposes a suite of
analytical techniques, that, when used in conjunction with each other,
provides a thorough characterization of the material.
Introduction
The formal defnition of nano particle is a discrete entity that has a
dimension of 100 nm or less. A nano-scaled material, on the other
hand, may be comprised of nano-scaled structures, though the
physical dimensions of the material may exceed 100 nm. A spray-dried
agglomerate of nano-sized particles would be described as nano-
scaled. For pharmaceutical applications, one does not resort to the use
of nano materials unless there is a therapeutic driving force to do so.
If the compound is poorly soluble in-vivo or if the molecule is highly
potent, and the desired presentation of the drug product is a tablet,
capsule, or suspension, the decision may be implemented to produce
the active pharmaceutical ingredient as a nano material.
The actual form of the nano materials covers a number of technologies,
from actual nanocrystals of API, to emulsions, to liposomes. An
excellent review of the current technology can be found in Ref.
[1]. Regardless of the formulation or carrier, once this size range is
implemented, the drug product performance is intimately connected
to the physical properties of the active pharmaceutical ingredient.
Physical characterization becomes a crucial part of the drug substance
and drug product control strategy.

January/February 2011 |

| 31

PArtIClE sIzIng
Physical Characterization techniques
Sample Preparation
To obtain accurate and meaningful particle size data, the challenge
for nano materials lies in the sample preparation. Because the
material is so small and so surface active, generally accepted
scientifc practices can often end up producing artifacts, or altering
the sample during measurement.
Toxicity concerns for highly potent nanocrystals suggest that the
material should not exist as a dry powder, but can be more safely
handled dispersed in a liquid. To prevent particle aggregation, either
the pH can be adjusted, or surfactants can be used. Solubility of drug
substance particles, however, is often pH dependent; therefore, if the
pH is adjusted to provide adequate particle dispersion, the material
could dissolve. The same phenomenon can occur with surfactants,
where the particles can be solubilized by their addition.
If the particles are not solubilized by the use of surfactants, then for some
characterization techniques, the adsorbed layer of surfactant will skew
the particle size, making the particles appear larger. This, however, may
be relevant to the performance of the drug, and such measurements
can be valuable in predicting in vivo performance [2]. Factors such as
these must be considered when interpreting physical characterization
data for nano materials in pharmaceutical applications.
Particle Size Measurement Via Dynamic Light Scattering
Laser difraction, the dominant platform for sizing most pharmaceutical
powders, is an ensemble technique (i.e. measures many particles
rather than a single particle measurement) that has a lower size limit of
approximately 0.1 m. To measure objects below that limit, dynamic
light scattering (DLS), also known as photon correlation spectroscopy
(PCS), is one of the few ensemble techniques that can be employed in
this size range (0.005 1 m). The technique is well established and
several commercial platforms are available. The theory and application
of this technique are contained in refs. [3-5].
Stable, well-dispersed particles are placed in the sample cell, where
Brownian motion causes the particles to move randomly in the
suspension. A laser beam passes through the sample cell and is
scattered by the particles. The randomly changing difraction pattern
is converted into a histogram of intensity vs. size. Note that this
data representation is not what one typically encounters, which is
frequency vs. time. For this reason, PCS should be used to measure an
average particle size rather than to produce a particle size distribution.
Figure 1 shows a schematic of a particle that has an adsorbed layer
of surfactant for particle dispersion. PCS measures a hydrodynamic
particle diameter; therefore, the adsorbed layer of surfactant will afect
the particle size measurement. The efective particle size is denoted
by the circumscribed blue circle. This is not necessarily an undesirable
occurrence. If this particle is being designed to pass through a specifc

32 |

|

January/February 2011
PArtIClE sIzIng

physiological barrier, such as the blood-brain barrier, or certain cell


membranes, the hydrodynamic measurement produced by PCS
would be extremely relevant. If the purpose of the measurement is to
measure the actual size of the particle itself, then the average particle
diameter would be larger than observed with other microscopic
techniques that would only image the denser particle.
Particle size measurement by Electron
microscopy
Since the inception of nano technology, electron microscopy has served
as the gold standard for measuring particle size and morphology.
Initially, scanning electron microscopes were incapable of imaging
nano-scaled objects; therefore transmission electron microscopy
was the only microscopic technique available. With the advent of
feld emission electron guns for scanning electron microscopes, this
scenario changed. Both instruments now ofer clear images of nano
materials. Figure 2 shows a scanning electron micrograph of drug-
carrying biodegradable nanoparticles comprised of a polysebacic acid
core and a shell of polyethylene glycol.
Scanning electron microscopy ofers a three-dimensional
representation of the particles. The gray scale produced in SEM
images, however, makes quantitative measurements on these images
using appropriate image analysis software very difcult because of the
lack of contrast provided.
Figure 3 shows a TEM image of nanoparticles of silicon dioxide (silica)
used for drug delivery. Because the electron beam passes directly
through the object in a TEM, electron difraction and fringes can be
observed. Rows of atoms can be observed in this image. Additionally,
there is much better contract in this image, allowing easier
quantifcation/measurement of features. Even with modern advances
in scanning electron microscopy, TEM exhibits much higher resolution.
For both transmission and scanning electron microscopy, the particles
must be isolated, and are analyzed in a high vacuum environment.
Both techniques are insensitive to adsorbed surface layers such as
surfactants that would be required to disperse the particles in a liquid
(typically aqueous) environment.
To obtain these high resolution images, in both instances, the sample
is bombarded with a very high energy electron beam. Prolonged
exposure of the particles to this beam can cause degradation. This
must be considered when opting for this type of analysis.
In summary, both PSC and electron microscopy provide information
on particle size. If at all possible, both techniques should be employed
for the size characterization of pharmaceutical nanoparticles. They
are complimentary orthogonal techniques that provide valuable
information.
Surface Area Measurements
Particle diameter is perhaps the most relevant data to collect for
scenarios where the physical size of the particle controls the in vivo
performance of the drug product. When nano-scaled formulations
Figure 1. Schematic of nano particle with adsorbed surfactant
layer. Particle size data obtained by PCS will also measure the
efect of the surfactant [12].
Figure 2. Scanning electron micrograph of drug-eluting
nanoparticles [13].:
Figure 3. Transmission electron micrograph of silica particles used
for drug delivery [14]

January/February 2011 |

| 33

PArtIClE sIzIng
are considered based upon the poor solubility of the drug substance,
surface area measurement provides equally meaningful data. It has
long been realized that surface area controls the dissolution of solid oral
dosage forms [6-11].
Because nanoparticles can easily pass through
the flters and frits found in commercial surface
area instruments, either the sample must be
exposed to vacuum very slowly, or the material
should be presented in an agglomerated form
(i.e. nano-scaled). If the nanoparticles are
contained in aggregates, a two-tiered strategy
for characterization could be adopted, whereby
the particle size measurement would measure
aggregate size, and surface area measurements
would provide an indication of the nano
particle size. Often, high energy sonication is
employed in an attempt to attain the primary
particle size. This is not a preferred choice.
The data obtained is often an indication of the
degree of sonication, rather than the primary
particle size.
Isoelectric Point and Zeta Potential
Isoelectric point and zeta potential
measurements are routinely collected data
by practitioners working with inorganic sub-
micron powders, nano particles, or colloidal
systems. The origin of these measurements is
found in the pioneering work performed by
Stern to describe the interaction of particles
in liquids. For nano particles such as proteins,
liposomes and nano crystals with low
isoelectric points, i.e. with low surface charges,
the nano material will easily focculate in the
suspension, creating an undesirable scenario.
For particles with a high zeta potential (either positive or negative), all
of the particles will have a high surface charge, thereby repelling each
other in the liquid and maintaining a discrete identity. The isoelectric
point of a particle in a liquid is the pH at which the surface charge
on the particle is zero, which is usually avoided in pharmaceutical
nanoparticulate applications. (For waste water treatment, the water
may be pH adjusted to achieve the isoelectric point, thereby causing
harmful particulates to focculate and allow them to be removed via
fltration). Figure 4 shows the classic diagram used to describe the
layers that form around a particle in a liquid
There are two analytical instrument platforms used to measure zeta
potential: electrophoretic and electroacoustic. With electrophoresis,
suspended particles are placed between the plates of a capacitor, and
the potential across the plates is scanned (typically from -25 to 25
mV). The particle motion is measured as a function of potential using
many of the same principles employed in dynamic light scattering
measurements. (Instruments are available that perform both particle
size measurement via DLS and zeta potential measurements.) With
electrophoretic measurements, the particles must be small enough
to remain suspended without the introduction of any additional
energy or agitation (stirring, pumping, sonication, etc.) Additionally,
the suspension must be suitably dilute to permit individual particle
motion detection by the laser.
Figure 4. Diagram illustrating particle surface charge in a liquid as
a function of distance from the surface of the particle [15].

34 |

|

January/February 2011
PArtIClE sIzIng

Electroacoustic instruments require more concentrated suspensions,


and the suspension can be stirred during the measurement. With
this technique, ultrasonic bursts of energy are pulsed through a
suspension, causing charged particles to move. This motion generates
an electrical charge, which is monitored as a function of the ultrasonic
energy burst. The sample can be stirred because the duration of the
pulse is so small; particle motion created by stirring is negligible.
Highly concentrated suspensions (up to 50 vol. %) can be measured
used this technique.
If the zeta potential of the nano material in its nascent state is
insufcient to prevent focculation, pH adjustments can be made
or surfactants can be added. The same limitations and cautions
described in Sample Preparation, however, are applicable here; pH
changes and the addition of surfactants can easily solubilize the
nano material.
Te need for a rigorous Physical
Control strategy
The mandate for any commercial drug product is to deliver safe
and efcacious treatment for human illness. All pharmaceutical
companies are required, by law, to demonstrate that they understand
the therapeutic action of the medicine, and they can reproducibly
manufacture the material to the same (or better) level of quality over
time. Physical and chemical tests are performed in a rigorous fashion
before any medicine can be released for human use. Such tests
include chemical tests for purity and potency, dissolution behavior,
moisture content; and often physical tests such as particle size (laser
difraction, sieve testing, etc.) and perhaps specifc surface area. For
drug product performance, typically dissolution testing is heavily
relied upon because this testing most closely imitates how the product
will perform in vivo, provided the drug product manufacturing process
is robust.
For nano materials in the pharmaceutical industry, the need for this
control strategy is even greater, specifcally for the physical properties of
the drug substance. The performance of the medicine is directly linked
to these properties. These properties will maintain themselves only
if the formulation is shown to be stable over the shelf life of the drug
product. It is quite likely traditional CMC strategies will be augmented
or changed to meet the more demanding needs of nano medicines, and
more sophisticated physical tests will be required in the development of
these compounds. Some of these techniques may also be used as quality
control tests to release the medicine prior to human consumption.
This additional testing may require pharmaceutical companies to develop
physical reference stands for nano medicines, much in the same way
reference standards are used for potency and related substance testing
today. Because drug product performance is linked to the drug substance
particle size in the formulation, reference standards may be required for
some physical property testing.
summary
Nano medicines ofer unique therapeutic advantages over their
traditional counterparts by exhibiting unique properties such
as enhanced solubility, selective cell targeting, and the ability to
pass certain biological barriers and membranes. This enhanced
performance/capability is linked directly to the physical properties
of the material. No single test can provide an adequate description
of the material. Great care must be taken to ensure that the sample
preparation steps do not alter the material. Tests not used routinely
in the pharmaceutical industry may provide the appropriate level of
scrutiny to interrogate the material. It may become necessary to use
some of these new techniques for quality control testing. Reference
materials could be necessary to ensure that physical testing data are
consistent and reliable for a given nano material.
references
1. Pathak, Y. and D. Thassu, eds. Drug Delivery Nanoparticles
Formulation and Characterization. 1 ed. Drugs and the
Pharmaceutical Sciences, ed. J. Swarbrick. Vol. 191. 2009, Informa
Healthcare: London. 394.
2. Banker, G.S. and C.T. Rhodes, eds. Modern Pharaceutics. 4 ed. Drugs
and the Pharmaceutical Sciences, ed. J. Swarbrick. Vol. 121. 2002,
Marcek Dekker: London. 838.
3. Xu, R., Particle Characterization: Light Scattering Methods. 2000,
Dordrecht: Kluwer Academis Publishers. 397.
4. Merkus, H., Partucle Size Measurements: Fundamentals, Practice Quality.
Particle Technology Series. Vol. 17. 2009, New York: Springer. 532.
5. ISO, 13321-1: Particle Size Analysis -- Photon Correleation
Spectroscopy. 1996, International Standards Organization: Geneva.
6. Iacocca, R.G., Physical Characterization Tests for API used in Low Dose
Formulations, in Formulation and analytical development for low-
dose oral drug products, J. Zheng, Editor. 2009, Wiley & Sons: New
York. p. 309-324.
7. Iacocca, R.G., C.L. Burcham, and L. Hilden, Particle engineering: A
strategy for drug substance physical property control during small
molecule development. J Pharm Sci, 2010. 99(1): p. 51-75.
8. Fukunaka, T., et al., Dissolution characteristics of cylindrical particles and
tablets. International Journal of Pharmaceutics, 2006. 310(1-2): p. 146.
9. Horter, D. and J.B. Dressman, Influence of physicochemical properties
on dissolution of drugs in the gastrointestinal tract. Advanced Drug
Delivery Reviews, 1997. 25(1): p. 3-14.
10. Thomas Schreiner, U.F.S.H.L., Immediate drug release from solid oral
dosage forms. Journal of Pharmaceutical Sciences, 2005. 94(1): p. 120-133.
11. Jinno, J.-i., et al., Effect of particle size reduction on dissolution and
oral absorption of a poorly water-soluble drug, cilostazol, in beagle
dogs. Journal of Controlled Release, 2006. 111(1-2): p. 56.
12. courtesy of D. Shekhawat
13. http://www.futurity.org/health-medicine/drug-toting-mucus-busting-
nanoparticles, Ben Tang and Mark Koontz/Johns Hopkins University
14. http://en.wikipedia.org/wiki/File:Mesopourus_silica_closeup.jpg
15. http://en.wikipedia.org/wiki/Double_layer_(interfacial)

January/February 2011 |

| 35

PArtIClE sIzIng
Author Biography
Ronald Iacocca received his B. S., M.S. and Ph.D. in
Materials Engineering from Rensselaer Polytechnic
Institute (RPI). For nine years prior to joining Lilly, he
was a faculty member in the Department of Engineering
Science and Mechanics at The Pennsylvania State
University. In November 2000, Ron joined Eli Lilly as a research scientist in
Physical and Structural Characterization, Pharmaceutical Product Research
and Development. Currently, he is a Senior Research Advisor in Analytical
Sciences Research and Development, and is team leader of the Materials
Science/Physical Characterization team. He has authored/co-authored
over 65 journal articles, book chapters, and review articles, and has been
granted 4 patents. He is a member of ASM International, ASTM, and ISO.
In 2005, he was selected as one of ninety world-wide experts by the Bill and
Melinda Gates Foundation to participate in the road-mapping initiative for
the development of a world-wide malaria vaccine.
Dr. Iacocca also served as an adjunct professor in the Department of
Industrial and Physical Pharmacy at Purdue University from 2006-2009,
and serves as a member of the editorial advisory board for American
Pharmaceutical Review, as a reviewer for the Journal of Pharmaceutical
Sciences, as a key reader for Metallurgical and Materials Transactions A,
and is head of the working group on laser difraction for the International
Standards Organization (Working Group 6, ISO TC24/SC4). In 2010, he was
elected to the USP to serve on the Physical Analysis Expert Committee
Many pharmaceuticals, such as monoclonal antibodies are produced
from genetically engineered mammalian cell lines. Purity with regard to
undesired host-cell proteins (HCP) of fnal product is essential for product
safety. Process development by varying upstream conditions and analyze
the effects on downstream processing is needed for optimal yield and
purity. We have characterized HCP patterns in purifcation of monoclonal
antibodies from CHO cells by using 2-D Fluorescence Difference Gel
Electrophoresis (2-D DIGE).
2-D DIGE technology with two different samples and a pooled internal
standard per gel pre-labeled with CyDye DIGE Fluor minimal Dyes,
detects differences in protein abundance. Experimental variation is
virtually eliminated and the quantitative data is very reliable.
Differences in protein expression between culture supernatants grown
with a set of altered media compositions were analyzed. Also, differences
in the HCP patterns of MabSelectSuRe eluent fractions were analyzed.
The results were related to yield of target protein and HCP levels obtained
with ELISA assay.
Presents this interactive webcast
Characterization of host cell protein
patterns using 2-D DIGE
Monday, March 21
3pm CET /10am EDT
Register online at:
http://tinyurl.com/4bo7usx
Speaker:
Maria Winkvist, Ph.D.
GE Healthcare, Life Sciences, Sweden
Maria Winkvist trained as a cellular and molecular
biologist at The Ludwig Institute for cancer research in
Uppsala, Uppsala University in Sweden, where she
received a Ph.D. in Medicine 2007. Her research was
focused on transcription regulation and cell signalling.
In 2007, Maria Winkvist joined GE Healthcare, Sweden,
as a specialist for Biomolecular imaging. She is part of
the Scientifc Support group, responsible among other
things for application development and training both
internally and externally.
Moderated by: Rita Marouga

36 |

|

January/February 2011
rAmAn

Joanny Salvas
1
, Jean-Sbastien Simard
1
,
Ryan Gosselin, Ph.D.
2
& Nicolas Abatzoglou, Ph.D.
2

1
Process Analytical Science Group, Pfzer Montral
2
Universit de Sherbrooke, Department of Chemical & Biotechnological Engineering, Industrial
Research Chair Pfzer/UdeS on PAT in Pharmaceutical Engineering
E-mail: Nicolas.Abatzoglou@USherbrooke.ca
Introduction
Context
The use of PAT has spread throughout the pharmaceutical industry;
mainly because PAT makes it possible to gain useful process insight,
improve monitoring throughout the manufacturing steps, lower costs
associated with the use of wet-chemistry-based testing methods,
and increase overall efciency. They are tools that ft very well in the
QbD scheme for better process control and understanding.
Many of the PAT methods are based on MultiVariate Predictive Models
(MVPM), and the calibration of such model is a step crucial to the
success of the MVPM-based PAT method.
The current trends in MVPM calibration are aligned with the mantra
the more the better. This implies that using more calibration points,
with more samples, manufactured as closely as possible to the
commercial product, with more precise reference methods, will lead to
more precise, better performing MVPM-based PAT methods. Obviously,
it also leads to a costlier and more time consuming development
process, thus limiting the potential applications as well as lowering the
overall payback.
Available literature, to the authors knowledge, does not contain
studies in which the predictive ability of multivariate models
elaborated with various calibration approaches is evaluated as
function of the latter. This is true for both Raman spectroscopy and
many other spectroscopic techniques. Thus, this work has been
undertaken to understand the infuence that diferent calibration
parameters have on the fnal performance of predictive multivariate
models and contribute to optimize the calibration process to
minimize cost and maximize performances.
This study was conducted using data collected during an actual
PAT application development. Salvas et al [(3),(4)] reported on the
development of a PAT for the quantifcation of minerals in intact tablets,
using Raman spectroscopy. The development of this project created
the opportunity to study more thoroughly the calibration processes
of multivariate models. From this application, out of the four minerals
for which multivariate models were developed, two were selected to
be part of the investigation: one in high concentration and one in low
concentration. They both come from the same commercial product.
They are referred to as Mineral 1 and Mineral 2 throughout this article.
Calibration of Multivariate
Predictive Models: The Study
of Factors Infuencing the
Prediction Accuracy of Raman
Spectroscopy Applied to
Pharmaceutical Tablets

January/February 2011 |

| 37

rAmAn
scope and methodology
The development of an MVPM includes several critical activities, one
crucial being the sample preparation for model calibration. Common
practice dictates that samples must be prepared in such a way that
they are as close as possible to real samples (i.e. taken from the
production line). To insure this similarity with full-scale situations,
scaled-down lab- and pilot-equipment are often used. While this
makes it possible to obtain more representative calibration samples
that provide good coverage of the calibration span, it is a very time-
consuming and costly activity.
As suggested by the QbD approach for developing a new product
or a new method, in the present case several parameters were
selected to be part of the present study [(5)]. The monitored
response for each trial was taken to be the accuracy of the final
method.
Eight factors (parameters) were identifed and tested independently
on two data sets. The tests were guided by a design of experiments
(DoE), making it possible to optimize the total number of predictive
models that needed to be made, while retaining sufcient data to
allow minimal confounding within second-order factorial interactions.
Each run of the DoE represent a multivariate model that is calibrated
with data obtained with a diferent set of sample-manufacturing or
data-organizing parameters. Factors and their levels are detailed in
Table 1.
In this work, a batch of sample refers to a given set of samples
all manufactured under the same conditions and at the same
time, corresponding to one calibration point (one set of materials
concentration). This work required the manufacturing of 26 batches
for 13 calibration points with each 2 manufacturing protocols
(diferent tablet presses, same overall batch volume). Replicates refer
to individual samples taken from the same batch.
Table 1: Parameters and tested levels for the DoE
PARAMETER
(alternate notations)
LEVEL TESTED
Type of press used for sample manufacturing
(Factor A, aka Press)
Automatic (27-station press)
Manual (single-punch press)
Distribution of calibration points over the calibration span
(Factor B, aka Distrib.)
Evenly distributed
Concentrated around target
Number of calibration points
(Factor C, aka NbPts)
Low (half )
High (all)
Number of replicates within each calibration point
(Factor F, aka NbRep.)
Low (half)
High (all)
Use of commercial samples for model calibration
(Factor E, aka Prod.)
Present
Absent
Calibration algorithm used
(Factor D, aka Algo)
PCR
PLS
Reference data used for modeling
(Factor G, aka Meas.)
Reference method value
Experimental manufacturing values

38 |

|

January/February 2011
rAmAn

A 27-3 design obtained with generators E=ABC, F=BCD and G=ACD


allowed to obtain level IV resolution, leading to double interactions
being confounding with each other but not with the main efects.
Another useful particularity of this fractional design is that it can be
collapse[d] into either a full factorial or a fractional factorial in [many
subsets] [] of the original factors. In the event that 3 factors are
found to be non signifcant, the probability of being able to transform
the design in a full 24 design is high [(2)].
Samples and calibration datasets were prepared according to the DoE.
Specifcs regarding sample manufacturing and model preparation
are reported in a previous paper and master thesis: Salvas et al [(3),
(4)]. These details are not repeated here and are not mandatory for
interpretation of the present study.
results and Discussion
Summarized results obtained after execution of the DoE are presented
in Table 2. The response monitored for the ANOVA is the accuracy of
the models and it is expressed as a form of error: the absolute value of
the relative diference between the Raman and the reference values.
Reference values were obtained by Inductively Coupled Plasma-
Optical Emission Spectrometry (ICP-OES).
All following analyses are based on the results presented in Table 2.
Efects of individual factors were determined through two separate
analyses of variance (ANOVA): one for each Mineral dataset. Results
of ANOVA tests are summarized in a modifed Pareto chart, illustrated
in Figure 1. In this fgure, a bar that continues past the signifcance
limit (p = 0.05, dashed line) indicates that the associated factor, in
the associated ANOVA, was found to have a signifcant efect on the
response. For example, factor B (Distribution of calibration points) is
demonstrated to have a signifcant efect in the case of the ANOVA on
Mineral 2, but not on Mineral 1.
Figure 1: Modifed Pareto chart of ANOVA main efects
Figure 2: Plot of means Mineral 1
Figure 3: Plot of means Mineral 2
Table 2: Prediction results for DoE models
RUN
P
r
e
s
s

u
s
e
d
P
o
i
n
t

d
i
s
t
r
i
b
u
t
i
o
n
N
b

o
f

c
a
l
i
b
r
a
t
i
o
n

p
t
s
A
l
g
o
r
i
t
h
m
C
o
m
m
e
r
c
i
a
l

s
a
m
p
l
e
s
N
b

o
f

r
e
p
l
i
c
a
t
e
s
R
e
f
e
r
e
n
c
e

d
a
t
a
| RELATIVE DIFFERENCE |
MINERAL 1 MINERAL 2
% %
1 -1 -1 -1 -1 -1 -1 -1 7.34 7.82
2 +1 -1 -1 -1 +1 -1 +1 6.85 1.64
3 -1 +1 -1 -1 +1 +1 -1 4.23 6.92
4 +1 +1 -1 -1 -1 +1 +1 2.34 9.24
5 -1 -1 +1 -1 +1 +1 +1 4.13 55.93
6 +1 -1 +1 -1 -1 +1 -1 1.28 30.24
7 -1 +1 +1 -1 -1 -1 +1 3.75 45.78
8 +1 +1 +1 -1 +1 -1 -1 4.85 38.22
9 -1 -1 -1 +1 -1 +1 +1 8.27 6.09
10 +1 -1 -1 +1 +1 +1 -1 7.98 11.44
11 -1 +1 -1 +1 +1 -1 +1 2.55 6.36
12 +1 +1 -1 +1 -1 -1 -1 9.13 1.78
13 -1 -1 +1 +1 +1 -1 -1 1.63 79.23
14 +1 -1 +1 +1 -1 -1 +1 1.14 60.52
15 -1 +1 +1 +1 -1 +1 -1 1.79 46.99
16 +1 +1 +1 +1 +1 +1 +1 3.19 21.45

January/February 2011 |

| 39

rAmAn
signifcant Parameters: Press used (A)
Test results indicate that factor A has a signifcant efect on the
measured response in the case of Mineral 2 (low concentration), but
not with the Mineral 1 dataset.
The use of a single-punch manual press requires small-scale
repetitive movements, such as measuring a few grams of the mixed
powder and letting it slide in the compression matrix. Despite using
mitigating techniques (such as limiting vibrations
and transfers), the smaller-scale movements
and manipulations apparently still induced
more local heterogeneity than bigger-scale
automatic presses. These local heterogeneities
seem to be more detrimental when trying to
link bulk concentrations to sub-samples of
less-concentrated materials. It might be that
increasing sampling volume when acquiring the
Raman spectra could help mitigating this efect,
but this remains to be tested.
significant Parameters:
Distribution and number
of Calibration Points (B
and C)
Factor B is signifcant only for the high
concentration material (Mineral 1), while Factor C
is signifcant for both. Since some inconsistencies
in the data have been observed, several additional
tests and analyses were done regarding these two
factors (details available in (4)).
These additional tests have led to the hypothesis
that an uncontrolled parameter was most
probably afecting the response. Several attempts
at pinpointing this parameter fnally lead to
the conclusion that it was the population in the
subsets of points selected for a given run that
afected the response. Indeed, when the number
of calibration points used is the maximum
available (such as in run 5 for example), there is
no positive degree of liberty left when assembling
the calibration matrix: all sample batches are
used. However, if the required number of point is
lower than the maximum available, a choice must
be made regarding which calibration batches are
selected for model elaboration.
During this work, when establishing a sub-
dataset, the line of thought has been to 1) cover
the whole range and 2) include batches in such
manner as to have an even distribution of points
along the span. Thus, calibration matrices were prepared for each run,
under these constraints. But, with lower levels, several alternatives
could satisfy the constraints. For example, dataset containing mixes
3, 8, 10, 11 & 13 or mixes 3, 4, 7, 8 & 12 both satisfed the constraints.
Given this situation, and the fact that problems with some batches had
been spotted previously [(3), (4)], it was advisable to study the efect of
this choice on the response.
A new design of experiment was prepared in order to infrm or confrm
the hypothesis that, when using the same number of calibration
WITec GmbH, Ulm, Germany
phone +49 (0)731 140700, info@witec.de www.witec.de
Confocal . Raman . Fluorescence . AFM . SNOM
WITecs new True Surface Microscopy allows confocal
Raman imaging guided by surface topography. The
topographic coordinates obtained by an integrated
prolometer are used to perfectly follow the sample
surface in confocal Raman imaging mode.
The result is an image revealing chemical properties
at the surface of the sample, even if it is rough or
inclined.
Automated Raman-AFM System alpha500
with Attached Sensor for Prolometry
Topographic Raman Image
of a Pharmaceutical Tablet
True Surface
Microscopy
Topographic
Raman Imaging
NEW
PRODUCT
WITec Instruments Corp.
phone +1 865 984 4445, info@witec-Instruments.com
40 |

|

January/February 2011

rAmAn

points, changing the members included in the selection had an efect on the response. Several
subsets of 5 batches (i.e. calibration points) were prepared and used to obtain models, with an
equal number of replicates in each batch.
Figure 2 and Figure 3 illustrate that, depending on the group of points selected and for both
components studied, there is a signifcant efect on the response.
Under the light of this new data, it has become clear that the infuence of factors B and C
(distribution and number of calibration points) can be equally due to the selection of the points
amongst all that were available as well as the added number of batches themselves. This could
mean that, since it is impossible to foresee which batch will turn out to contain good variance
and thus improve the model calibration, disposing of a high number of calibration points
from which to choose an adequate sub-set for model calibration might reveal to be more of
an advantage than saving costs by reducing the number of available diferent batches. The
optimal level for factor C must hence be high, so the best selection can be made amongst
available batches, no matter if the points are equally distributed or not.
non signifcant Parameters
Within the framework of this study, the following factors do not appear to have signifcant efect
on the response, for neither the Mineral 1 nor the Mineral 2 datasets:
factor D (calibration algorithm used);
factor E (use of commercial samples in the calibration set);
factor F (number of replicates used per calibration point); and
factor G (type of reference measurement used in the calibration step).
The conclusion reached for factor D is in accordance with literature on the subject. It is indeed
widely agreed that both PLS and PCR algorithms can lead to a good model, but that, in general,
PLS will do it using fewer Principal Components (PC) [(1)].
No literature was found on the use of commercial samples in the calibration stage of an MVPM-
based PAT (factor E), but the results obtained are in accordance with the general calibration
procedure. As the goal is to produce samples that are as close as possible to the future ones
(commercial), it is not surprising that, when such goal is achieved, including the real commercial
samples in the calibration set does not make a statistically signifcant diference. In the case real
scale samples are difcult to obtain, the inclusion of commercial samples in the calibration set
might be an asset for fne-tuning the model, but this remains to be tested, as it was not part of
the present work.
The conclusions reached in the case of factor F are a pleasant surprise. It was expected, in
accordance with the general calibration procedure, that including more replicates, and thus
more variation in the calibration matrix, would allow the model to better perform in both the
validation stage and its routine use. It appears that, no matter how many replicates are included
in each calibration point, the response is not afected. It was expected that a decreasing
asymptote-like response-plot of prediction accuracy vs replicate number would be obtained
when increasing the number of replicates This would have suggested an optimal number of
replicates at the lowest error obtained with the least number of samples; it was not the case. The
results obtained can be explained by the fact that the commercial samples are manufactured
in such a manner that they are very uniform from batch to batch; thus, the inclusion of more
variation in the calibration stage may not be as crucial as in other cases where more variation will
be encountered in the application of the model. On the other hand, including more replicates in
the calibration stage may increase model robustness over time. This was not investigated in the
present study and should be kept in mind in future work.
Enwave
Raman Solutions for
PAT Success!
On-line Process
Laboratory Discovery
ulill our anltical
goals with better tools
Enwave Optronics, Inc.
Tel: (949) 955-0258
info@enwaveopt.com
www.enwaveopt.com
Handheld
Raman
ProRaman-I
ProRaman-L
Raw Material Release
Optronics

January/February 2011 |

| 41

rAmAn
The results obtained regarding factor G are the most surprising. It
was expected that a signifcant diference would be observed when
using reference methods rather than theoretical values for y-variables,
especially for low-concentration materials such as Mineral 2. This was
expected due to (1) the poorer quality of the samples developed
at lab-scale, (2) the low concentration values of the raw materials
and (3) the sub-sampling inherencies of spectroscopic techniques.
Indeed, because Mineral 2 is less concentrated, a slight error in the
manipulations should have had a higher impact on the accuracy of
the expected values associated with the samples. Moreover, the higher
variation of the sample-to-sample inhomogeneities in the probed-
region was expected to cause even more inaccuracies regarding the
expected concentration-values of each individual tablet and spectra.
Alternatively, a mineral with higher concentration was expected to
be distributed more homogeneously in each sample, thus decreasing
the impact of potential manipulation errors on the accuracy of the
theoretical values associated with the batch.
In light of the results obtained, the previous explanations must be
revisited. First, it can be noted that the reference measurements
have an error (or precision) of their own. Including the reference
measurements in the calibration stage includes their error in the
model. Theoretical values also have an error, but there is no a priori
knowledge as to which error is better, even though it was originally
postulated that the reference acquired with a compendial method
should de facto be. Training the model with the reference error was
expected to systematically give better result.
The mean diference between the theoretical and the reference values
was checked for the samples manufactured with the automatic press,
and it is of 1.9 and 1.2 % for Mineral 1 and 2, respectively. This deviation
is within the reference methods precision range, suggesting that the
diference might not be statistically signifcant. Under the light of this
information, the conclusions reached regarding the efect of factor G
make more sense. Both concentration measurements are apparently
equivalent, showing that the models are equally well-calibrated.
The diference between reference and theoretical values are greater
when dealing with samples manufactured using the manual press
(respectively 3.9 and 2.3 %), but still not big enough to have an impact
on the accuracy of the multivariate model. The diference is nonetheless
in agreement with the fact that factor A (press used) was found to have
a signifcant efect on the response.
Nevertheless, since manipulation errors are intrinsically various and
mostly not repeatable, it is possible that, while in this case they are
equivalent to that of the reference method, they might be higher
or lower in any other case. More work should hence be done on
that subject.
Raman solutions
for the pharmaceutical industry
email: ad.sci@horiba.com www.pharmaraman.com
Sometimes you have to look below the surface.
From fast imaging tools in drug discovery through
targeting bottlenecks in drug development and
providing new technology solutions for assuring drug
product quality in manufacturing, HORIBA Scientic
is continuously meeting new challenges
across the breadth of the industry.
Raman solutions- iceberg half page for APR.indd 1 2/4/2011 12:18:40 PM

42 |

|

January/February 2011
rAmAn

optimized Protocol Proposal


Conclusions reached regarding the efect of each factor on the accuracy
of the method allowed proposing an optimized development protocol
for MVPM-based PAT methods, as described in Table 3.
An automatic press should be used to produce a high number of evenly
distributed calibration points. A PLS regression algorithm should be
used to calibrate the predictive model, using theoretical y-variables as
a starting point. The number of replicates can be lowered signifcantly
if it makes it possible to gain time or reduce eforts and/or costs.
Using these sets of parameter settings should allow the development
of a reliable predictive model, at reduced cost and with an accelerated
schedule. This optimized method was tested in order to verify the
overall impact on performance by selecting the data in the calibration
matrix that would have been obtained if such a protocol had been
used. Three (3) more multivariate models were hence developed, one
for each of the monitorable minerals presented in Salvas et al [(3), (4)].
Prediction errors, calculated as the relative diference between the
claim and the measurement, are presented in Table 4. The diference
in precision does not exceed 2.5 %(absolute value) in the worst case
observed and would have allowed savings in direct invested man-
hours of approximately 30 % and overall development-cost, by 31 %.
Conclusion
This work has resulted in the recommendation of an optimized
development protocol for Process Analytical Technology based on
MultiVariate Predictive Models (MVPM-based PAT).
Beside the monetary savings the main advantage of these two changes
is that it allows cutting the estimated development time by two. This is
a tremendous advantage, given the always limited resources that can
be allocated to such projects. This time reduction allows the facility
to undertake more diferent projects at the same time and hence
multiply the potential gains and return on each system investment.
This work was based on a specifc PAT application (Raman spectroscopy
for content-monitoring of intact tablets). Further work is undergoing
with diferent applications (other spectroscopic techniques, other
types of products, other parameters) in order to pursue and challenge
these conclusions. Many other parameters could afect method
performance, but the present work has demonstrated that it is possible
and wishable to lower the burden of multivariate model calibration by
optimizing the development protocol.
Acknowledgments
The authors are indebted to Pfzer Montreal, The National Science &
Engineering Research Council (NSERC) of Canada and the Universit
de Sherbrooke for funding related with the project.
Anne-Marie Demers and the late Jasmin Groleau are thanked for their
precious help in preparing the many samples required for this work.
Ryan Gosselin and Guillaume Leonard are also kindly thanked for their
help in revising this article.
references
1. Esbensen, K.H. (2004.) Multivariate data analysis in practice. 5th edition, Esbjerg, Camo
Process AS, 598 p.
2. Montgomery, D.C (2001). Design and Analysis of Experiments. 5th edition, Wiley, New York, 684 p.
3. Salvas, J., Simard, J.-S., Abatzoglou, N. (2010a) Raman Spectroscopy to Analyze Intact
Pharmaceutical Tablets, American Pharmaceutical Review, April 2010, p. 46-53
4. Salvas, J. (2010b). Calibration of multivariate predictive models: the study of factors
infuencing the prediction ability of Raman spectroscopy applied to pharmaceutical tablets,
Master thesis, University of Sherbrooke, Canada.
5. United States. International Conference on Harmonisation (2008). ICH Harmonised
Tripartite Guideline Pharmaceutical Development Q8(R1). Current Step 4 version dated
13 November 2008, accessed online (http://www.ich.org/LOB/media/MEDIA4986.pdf )
September 01 2009, 28 p.
Table 3: Recommended levels for each tested factor
FACTOR
LEVEL
TESTED OPTIMAL RECOMMENDED
(A) Type of press used Automatic, Manual Automatic Automatic
(B) Distribution of calibration points Concentrated, Even Even Even
(C) Number of calibration points Ca: 3, 5, 6, 8, 12, 13
Mg: 4, 5, 6, 8, 9
High
number
High number
(D) Calibration algorithm used PCR, PLS - PLS
(E) Use of commercial samples Absent, Present - Most convenient
(F) Number of replicates 3, 5, 7, 10 - 5
(G) Type of reference used Theoretical, Reference - Theoretical
Table 4: Comparison of error for optimized models
MODELS MINERAL 1 MINERAL 2 MINERAL 3
FULL MINIMAL FULL MINIMAL FULL MINIMAL
% % % % % %
Prediction error 2.9 1.9 2.6 3.1 5.6 8
Diference
full vs minimal
-1.0 +0.5 +2.4
January/February 2011 |

| 43

rAmAn

Biography
Dr. Nicolas Abatzoglou is full professor and Chairman
of the Department of Chemical & Biotechnological
Engineering of the Universit de Sherbrooke (UdeS). He is
a specialist in Process Engineering involving particulate
systems in reactive and non-reactive environments.
He is the holder of the UdeS Pfzer Industrial Research Chair on Process
Analytical Technologies (PAT) in Pharmaceutical Engineering. He is co-
founder of the company Enerkem Technologies Inc., a spin-of of the
Universit de Sherbrooke in the feld of energy from renewable resources.
He has a career of many years at both the academic and industrial levels.
His professional experience as engineer spreads over the last twenty years.
He represented Canada at the International Energy Agency (Gasifcation
Task) from 1997-2001 and was the secretary of the Board of Directors
and the Executive Committee of the AQME (Association qubcoise pour
la matrise de lnergie) from 1996-2000. His production as a researcher
includes a hundred of publications in scientifc reviews, international
conferences, plenary and invited lectures, patents and a book chapter.
Jean-Sbastien Simard has a Master degree in
Chemical Engineering specialized in particulate systems
for direct compression from Universit de Sherbrooke,
Qubec, Canada. He is currently pursuing a MBA degree
at Universit Laval, Qubec, Canada. He has been with
Pfzer Canada for the last ten years, where he worked as a Product and
Process Development Scientist for the pharmaceutical processing unit.
For the last fve years, he has been responsible for the Process Analytical
Technology Development Group in the Technical Services. He has co-
authored many diferent presentations on particulate systems behavior,
Quality by Design and Process Analytical Technology applications. He
is also the Industrial Responsible of the UdeS/Pfzer Industrial Research
Chair on Process Analytical Technologies in Pharmaceutical Engineering.
Joanny Salvas has a bachelors degree in
biotechnological engineering. She has completed a
M.Sc.A. in chemical engineering with the Universit
de Sherbrooke, conducting her graduate work at
Pfzer Montreals facility (Canada), as part of a Chair
partnership. Her research aimed at optimizing the development protocol
of multivariate predictive models used as part of PAT methods. She
currently is a PAT Scientist at Pfzer Montreal, where she pursues several
projects with diferent technologies such as Raman spectroscopy
and Rapid Microbiological Methods. She is also leading projects for
international sites.
Dr. Ryan Gosselin is an assistant professor at
the Department of Chemical & Biotechnological
Engineering of the Universit de Sherbrooke, Canada. He
is a specialist in Process Engineering and on-line quality
control through the use of multivariate data analysis
and chemometrics. As a member of the Pfzer Industrial Research Chair
on Process Analytical Technologies (PAT) in Pharmaceutical Engineering,
his present work focuses mainly on issues relating to the production and
handling of non-reactive particulate systems.
Dont Burn Through
Your Money
Burn Through Your Work!
Our DSC Instrument features:
User-replaceable/corrosion resistant sensor to
minimize maintenance costs
TOPEM, the new multi-frequency modulated
temperature DSC technique from METTLER TOLEDO
High heating rates to reduce measurement time and
increase productivity
Reliable auto-sampler to maximize sample analysis
and throughput
www.mt.com/thermalexcellence

44 |

|

January/February 2011
DIssolutIon

Saeed A. Qureshi
Senior Research Scientist, Therapeutic Products Directorate,
Health Canada, Banting Research Centre
Email: saeed.qureshi@hc-sc.gc.ca
Limitations of Some
Commonly Described
Practices in Drug Dissolution
Testing and Suggestions to
Address These
Introduction
Dissolution tests are employed to establish the quality of drug
products, mostly tablets and capsules, based on in vitro drug release
characteristics of these products. In reality, a dissolution test may be
considered as a simple extraction step in a vessel with a stirrer. Most of
the commonly used apparatuses in this regard are known as paddle and
basket apparatuses, in which a round bottom vessel (1 L) containing a
stirrer referred to as paddle (an inverted T-shaped bar) or small wired
cage (known as basket), respectively, are used. These apparatuses are
very well recognized and used around the world with the acceptance
of regulatory and standard setting organizations. Detailed descriptions
about these apparatuses may be found in any of the most commonly
followed pharmacopeias such as United States Pharmacopeia (USP) [1].
As noted above, drug dissolution testing is a relatively simple
technique, however, serious concerns and problems are often
reported in the literature about it [2-5]. These reported problems
often relate to: (1) failing of the performance evaluations of the
apparatuses (calibration) and/or products; (2) lack of establishing
the link between in vitro dissolution results and in vivo results,
commonly referred to as in vitro-in vivo correlations or IVIVC; (3) lack
of objectivity in setting or selecting experimental conditions for
product evaluations (4) setting unreasonably wide tolerances based
on complex and convoluted rationales. These wide spread concerns
result in frustrations, within both regulatory and manufacturing
environments, where objectivity and reliability of an analytical
technique is of critical importance for establishing the standards for
the assessment of quality of the drug products.
With such frustrations, it has been suggested that the dependence on
drug dissolution testing should be eliminated [6]. As drug dissolution
testing is an important and relevant step, the question obviously
should be that what went wrong in the practice of drug dissolution
testing rather than removal of the test that is mandatory [7]. This
article will present a discussion as to why there are such concerns and
describe some solutions to address these concerns.

46 |

|

January/February 2011
DIssolutIon

Background Information objective


of Drug Dissolution testing
The quality of an oral drug product (tablet and capsule) is based on
the fact that the drug will be released from a product in a predictable
and reproducible manner and dissolved in the fuid present in the
human gastrointestinal (GI) tract, in particular, small intestine. Thus,
this in vivo drug dissolution step, also interchangeably referred to as
drug release, becomes a critical step for developing a product and
later assessing its quality.
A drug dissolution test, or simply dissolution test, is conducted to
mimic the above described in vivo dissolution behavior of the drug in
vitro. It cannot be emphasized enough to highlight the fact that a drug
dissolution test is a test to evaluate in vivo dissolution behavior of a
drug product. There is no other objective or rationale for conducting
this test. However, there is a common practice for describing and using
the dissolution test, without its stated link to in vivo, for establishing
batch to batch consistency of the product and this is referred to as a
quality control (QC) test.
Unfortunately, this appears to be a misconception about the practice
of drug dissolution testing and leads to current problems and concerns
about the technique. If the link to the in vivo behavior is ignored,
then the obvious question would be: what parameter/characteristic,
or consistency thereof, a dissolution test refects. In addition, what
would be the basis of selecting experimental conditions to conduct
a dissolution test? It is, therefore, important and critical to note that
the only purpose or objective of dissolution testing is to assess the
in vivo release behavior of a product. Keeping this objective in mind
should also help and guide in defning experimental conditions for
dissolution testing.
Evaluating and relating to In Vivo
Dissolution Behavior
Once the objective is set, as described in the previous section, then
the question should be how one would relate the in vitro results to
in vivo dissolution characteristics? This question should be answered
in two parts. The frst part addresses the fact that the dissolution test
be conducted by mimicking, not necessarily duplicating, the in vivo
or intestinal environment. The second aspect should be a comparison
of the in vitro results to the in vivo. The discussion regarding the frst
answer is provided in the following section, however, discussion on
the second aspect is provided in this section.
In this regard, the most commonly reported practice is that of
developing or establishing an in vitro-in vivo co-relationship or IVIVC.
The commonly cited defnition of IVIVC from the US FDA guidance
document is: It defnes IVIVC as a predictive mathematical model
describing the relationship between an in vitro property of a dosage
form (usually the rate or extent of drug dissolution or release) and a
relevant in vivo response, e.g., plasma drug concentration or amount
of drug absorbed [8]. The preferred or desired IVIVC outcome is of
level A which implies comparing point-by-point (time-by-time)
in vitro dissolution results with in vivo dissolution results extracted
from drug concentration-time profles. Conversely, by comparing
predicted drug concentration-time profles obtained from in vitro
dissolution results with the actual drug concentration-times profles
obtained from bioavailability/bioequivalence (BA/BE) studies. Apart
from lack of clarity on the mechanics (procedure) of obtaining in vivo
dissolution results or deriving blood levels from in vitro dissolution
results, suggested IVIVC practices appear to have serious limitations.
For example: As the name IVIVC implies that one is required to develop
relationships between in vitro and in vivo results. However, in practice,
conducting a dissolution test is never meant for establishing such a
relationship as this relationship is considered to be always present. In
fact, existence of this relationship (IVIVC) forms the basis of conducting
of a dissolution test. It appears that there is serious confusion in the
literature in this regard. The purpose of dissolution testing should be
or has always been to evaluate characteristics (quality) of the product,
based on the underlying principle that a dissolution test relates well to
the products in vivo dissolution characteristics.
Even when such a relationship is developed, as current practices require,
by conducting both in vitro (dissolution) and in vivo BA/BE studies
using single product or multiple products with diferent formulation/
manufacturing variations, the question becomes what did one achieve
from this practice. If one gets a perfect correlation then it would show
that dissolution results are capable of predicting in vivo results. Is it not
that a dissolution test is conducted based on this principle in the frst
place, i.e., a dissolution test is conducted to refect potential in vivo
behavior of a drug product. Then, why does the development of IVIVC
have to be repeated with every drug and product?
There is another major faw in the current practices of IVIVC. These
practices of so called IVIVC seek a matching (rather than relationship)
by adjusting experimental conditions so that in vitro results would
match the in vivo outcome. Thus, in reality, the practice of IVIVC has
become a practice of searching test/experimental conditions to match
in vitro dissolution results of test product(s) to the in vivo results.
Furthermore, such successful IVIVC outcomes, which are rare, are
hardly used in practice to evaluate the quality of drug products. The
procedures which are used for the evaluation of the quality of products
(such as pharmacopeial tests) are generally not based on these IVIVC
evaluations. This obviously adds to the frustrations as to why IVIVC
studies are to be conducted when it may not be useful in assessing the
quality of the product.
The question would then be, what is the intended purpose of the
IVIVC practice? The intended purpose of the practice of IVIVC is not
to develop (co)- relationship but to predict, more accurately estimate,
a potential in vivo outcome. The in vivo outcome which is often used
in this regard is drug concentration-time (C-t) profles obtained from
the BA/BE studies. Therefore, the objective of any dissolution testing
must be to estimate and evaluate the C-t profles. A detailed discussion
on the procedural detail about developing such C-t profles and their
evaluations are beyond the scope of this article. Readers are referred
to the literature on this subject, where necessary concepts and
methodologies, in this regards, are provided in detail [9, 10].

January/February 2011 |

| 47

DIssolutIon
Choice of Experimental Conditions
As dissolution test are conducted to evaluate potential drug release in
vivo i.e., in the GI tract, choice of experimental conditions are, therefore,
dictated by the physiological environment. Basically there are three
variants which are usually considered in this regard: (1) temperature,
which is 37 C refecting body temperature; (2) GI tract fuid which
is refected by water or aqueous solutions (bufers) having pH in the
range of 5 to 7. If a drug is not expected to dissolve in water or bufers
then a small amount of solubilizing agent may be added to enhance
the solubility in the aqueous phase; (3) a mixing mechanism which
is achieved by using a stirrer at a slow rotation speed. In short, water
alone as a dissolution medium, or with small amount of solubilizing
agent if the drug is of low aqueous solubility, maintained at 37 C with
a stirrer at low rotation speed of 25 rpm may be used for testing of
the majority of drug products [11]. It is to be noted that experimental
conditions are derived from the physiological environment which
remains the same from product to product thus these have to be
product independent. However, a quick review of the literature shows
that most experimental conditions, except temperature, are product
dependent. Conducting dissolution studies using product dependent
experimental conditions clearly negate the basic requirement of
the testing. This creates a serious concern about the relevancy and
credibility of current practices of dissolution testing, thus results
obtained from dissolution testing would be of questionable merit.
At present there are two sets of variants in selecting experimental
conditions for dissolution testing; media and apparatuses or stirrers.
Commonly dissolution results are dependent on these two variants.
In most cases, two types of apparatuses are used i.e., paddle and
basket. These two types of apparatuses are similar in make and
operation, expect for the stirring rods (or spindles). It is very well
established, based on reports published in the literature, that these
apparatuses are inherently fawed for dissolution testing because
of poor hydrodynamics (mixing/stirring) within the vessels [2-4].
This fawed hydrodynamics results in serious defciencies; such that
the stirring provides limited product/medium interaction as well as
creates unstirred and stagnant pockets. The physiological relevance
of these apparatuses would thus be questionable as the intestinal
environment provides thorough mixing and no stagnant pockets.
Secondly, again based on the poor hydrodynamic characteristics,
it has clearly been demonstrated that these apparatuses provide
highly variable and unpredictable dissolution results unrelated to
a products characteristics. Therefore, results obtained using these
apparatuses will always be suspect and of limited use. There have been
numerous attempts and suggestions for improving the behaviors of
the apparatus by tightening specifcations [12], but with little success
as the issue does not appear to be with the specifcations (tight or
relaxed) but the apparatuses themselves.
Furthermore, as the cause of the problems is poor hydrodynamics
within vessel using paddles and baskets, then, it may not be possible
www.sotax.com sotaxusa@sotax.com sotaxusa@sotax.com
SOTAX Corporation 68A Elm Street Hopkinton MA 01748 USA Toll Free 1 888 SOTAXUS E-mail sotaxusa@sotax.com
Tech Support sotax-techsupport@sotax.com Tech Support Hotline 1 508 544 4040
Fully Automated Dissolution System
What SOTAX Automation customers are saying?
I was asked to perform 80 method development lots
including multiple rpms and buffers. Without SOTAX
automation it would have taken me weeks to perform.
It took me 4 days including data reporting
We have estimated a cost savings of over $250,000/
year for our 3 highest volume products in our QC lab
The method included a time consuming full media
change that took all day including analysis. With SOTAX
automation, we reduced our testing time by 60%
We have noticed a signicant reduction in lab errors
and investigations due to our SOTAX automation
system
SOTAX has provided us excellent service and technical
recommendations and solutions
Are you ready to start saving? Contact your local
SOTAX ofce to schedule your demonstration.

48 |

|

January/February 2011
DIssolutIon

to make an appropriate choice of a dissolution medium using these


apparatuses. The dissolution results obtained thus will always include
high variability and unpredictability of the apparatuses. Unfortunately,
instead of focusing on the issues of the apparatuses, there has been
a tradition of supporting the use of paddle and basket apparatuses
with weak rationales. The continued use of the paddle and basket
apparatuses appears to be the major impediment of addressing the
problems in developing appropriate dissolution tests [13].
looking to the Future
The obvious question would be as to how these issues may be
addressed. Obviously frst and foremost is the need for recognition of
the fact that unfortunately the recommended apparatuses (paddle and
basket) are not appropriate for their desired purpose. There is strong
experimental evidence in the literature regarding the defciencies [5]
as well as suggested solutions to address these [14]. However, there
appears to be a lag in recognizing these developments. It is hoped
that these new developments will provide impetus to re-evaluate the
future use of paddle and basket apparatuses.
On the other hand, to accommodate the continued use of these
apparatuses, at present, it has become a common practice to
select arbitrary experimental conditions such as apparatus, rpm,
dissolution medium etc. to achieve preconceived or expected
dissolution characteristics of a product. The current practices of
dissolution testing are therefore, in fact exercises of selecting/
defning experimental conditions to obtain expected dissolution
behavior rather than determining dissolution characteristics of the
products. Hence, it would be safe to conclude that with the current
recommended practices of dissolution testing one never determines
the drug release (dissolution) characteristics of product.
In resolving the issue, it appears that there is a need for clearly defning
and agreeing to the role of dissolution testing (evaluation of in vivo
drug release) with an objective endpoint (developing C-t profles).
Such an objective and end point will facilitate the development/use
of appropriate apparatuses and associated experimental conditions.
One of the possible ways of achieving such an objective is through
the availability of a reference product with known in vivo drug
release characteristics. Such a reference product should be used
in establishing the appropriateness of apparatuses and related
experimental conditions. The use of such validated apparatuses and
experimental conditions should be extended for other products. It
is ironic that the drug dissolution community has been working for
the past 3/4 decades and is expected to continue to work without
a reference product. It is highly unlikely, if not impossible, that one
will be able to get useful results from a technique/apparatus which
has not be validated for its claimed propose. It is a critical defciency
which requires urgent attention.
In the absence of such a reference product, as well as for generating
data towards developing a reference product, one may establish
appropriateness of an apparatus and associated experimental
conditions based on relative dissolution testing. The relative dissolution
testing may be described as determining drug dissolution (release)
characteristics of two products of the same drug (active ingredient) but
having two diferent known drug release characteristics in vivo such
as IR and ER products. The dissolution test conditions should refect
a physiological environment and must be such that while providing
diferent release patterns, fast for IR and slow for ER product, provide
complete dissolution to occur within the suggested dosing interval
for the drug products. Once such a set of experimental conditions is
established, this may be considered as refecting/simulating in vivo
environment and then be used for other test products. It is to be
noted that using experimental conditions which are not observed in
vivo, such as de-aeration of dissolution medium, use of sinkers etc., be
avoided as these may invalidate the testing.
In conclusion, it may be argued that most of the defciencies/
problems of current practices of dissolution may be related to poor
hydrodynamics within the paddle and basket apparatuses which also
lack relevance to physiological environment. The dissolution testing
may signifcantly be improved if its role may clearly and objectively
be established that the tests are to be conducted only to refect in vivo
dissolution characteristics of a product. This clarity of objective will
provide an improved basis for selecting appropriate apparatuses and
associated experimental conditions. In addition, such an objective
will also reduce signifcant work load by eliminating requirements
of repeated IVIVC developments and other physiologically non-
relevant testing.
Disclaimer: Views expressed here are for scientifc discussion purposes
only and may not be refective of opinions and policies of my employer
(Health Canada).
references
1. USP General Chapter on Dissolution <711>. United States
Pharmacopeia and National Formulary; United States Pharmacopeial
Convention, Inc.: Rockville, MD, 2008. p.267-274.
2. Baxter JL, Kukura J, Muzzio FJ. Hydrodynamics-induced variability in
the USP apparatus II dissolution test. Int J Pharm 2005;292:17-28.
3. Healy AM, McCarthy LG, Gallagher KM et al. Sensitivity of dissolution
rate to location in the paddle dissolution apparatus. J Pharm
Pharmacol 2002;54:441-4.
4. Qureshi SA, McGilveray IJ. Typical variability in drug dissolution
testing: Study with USP and FDA calibrator tablets and a marketed
drug (glibenclamide) product. Eur J Pharm Sci 1999;7:249-258.
5. Qureshi SA. A new crescent-shaped spindle for drug dissolution
testing - but why a new spindle? Dissolution Technol 2004;11:13-18.
6. DSouza1, S.S., Lozano, R., Mayock, S., and Gray, V. AAPS Workshop
on the Role of Dissolution in QbD and Drug Product Life Cycle: A
Commentary. Dissolution Technol. 2010, 17 (4), 41-45.
7. Tong, C., Lozano, R., Mao, Y., Mirza, T., Lbenberg, R., Nickerson, B., Gray,
V., and Wang, Q. The Value of In Vitro Dissolution in Drug Development
A Position Paper from the AAPS In Vitro Release and Dissolution Focus
Group. Pharmaceutical Technology 33 (4), 52-64 (2009).
8. US FDA Guidance for Industry (1997): Extended Release Oral
Dosage Forms: Development, Evaluation, and Application of In
Vitro/In Vivo Correlations. http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/UCM070239.
pdf (Accessed December 29, 2010).

January/February 2011 |

| 49

DIssolutIon
9. Qureshi, S.A. In vitro-in vivo correlation (IVIVC) and determining
drug concentrations in blood from dissolution testing A simple and
practical approach. The Open Drug Delivery Journal, 2010, 4, 38-47.
(Link). (Accessed December 30, 2010).
10. Qureshi, S.A. Determining blood concentration-time (C-t)
profiles from in vitro dissolution results and product evaluation
carbamazepine. http://www.drug-dissolution-testing.com/?p=601
(Accessed December 30, 2010).
11. Qureshi SA. Drug dissolution testing: Selecting a dissolution medium
for solid oral products. Am Pharm Rev 2009;12:18-23.
12. Gray, V.A. Identifying Sources of Error and Variability in Dissolution
Calibration and Sample Testing. Am. Pharmaceutical Reviews. 5:2
(2002) 8-13.
13. Gray, V., Kelly, G., Xia, M., Butler, C., Thomas, S. and Mayock. S. The
Science of USP 1 and 2 Dissolution: Present Challenges and Future
Relevance. Pharmaceutical Research, 26:6, 2009, 1298-1302.
14. Qureshi SA. A Crescent-shaped Spindle for Improved Dissolution
Testing. Pharmeuropa Bio & Scientific Notes, 1:2009: 55-66.
Author Biography
Dr. Qureshi is a senior research scientist in the Therapeutic Products
Directorate, Health Products and Food Branch, Ottawa, Canada.
His main area of research involves the assessment of drug release
characteristics, both in vitro and in vivo, of oral and dermal products. He
has published more than 40 papers, including a chapter in Encyclopedia
of Pharmaceutical Technology as well as made numerous national
and international presentations in the areas of drug dissolution
testing, analytical chemistry, pharmacokinetics, bioavailability and
bioequivalence. Dr. Qureshi, moderates and is a frequent contributor to a
blog on the subject (www.drug-dissolution-testing.com).
Reach your target.
If you need authoritative, unbiased literature to enhance your product or service,
what better way than to order reprints of one of the articles in this or any issue of
American Pharmaceutical Review. Professionally presented with an advertisement
or simply your contact details, reprints are a great way to showcase your company.
For more information about purchasing reprints,
contact us at 317-816-8787 or at info@russpub.com

50 |

|

January/February 2011
mICroBIology

Kalavati Suvarna, Ph.D, Anastasia Lolas, M.S.


Patricia Hughes, Ph.D. & Richard L Friedman, M.S.
Biotechnology Manufacturing Team, Division of Manufacturing and Product Quality,
Ofce of Compliance, Center for Drug Evaluation and Research, Food and Drug Administration
Email: kalavati.suvarna@fda.hhs.gov.
Case Studies of Microbial
Contamination in Biologic
Product Manufacturing
Abstract
The manufacture of biologic products is a complex process and
requires the use of living cells. These processes and products are prone
to contamination by adventitious agents such as bacteria, fungi and
viruses. Microbial contaminations have a huge impact on biologic
product manufacture as they introduce product variability and can
cause loss of potency due to degradation or modifcation of product
by microbial enzymes, changes in impurity profles, and an increase
in the levels of bacterial endotoxins. In addition, the investigations of
microbial contaminations can result in lengthy shutdown periods and
delays in manufacturing operations that in turn, may sometimes result
in shortages of essential drug products. Strict microbial production
controls are essential to ensure the manufacture of a drug product with
consistent quality. This article discusses elements of a microbial control
strategy, recent cases of microbial contamination in specifed biologic
products, the need to perform risk assessments on a periodic basis, and
additional areas of improvement in the management of risks.
Introduction
Biologic products are manufactured using living cells such as bacteria,
yeast, and mammalian cells. These include specifed biologics such as
monoclonal antibodies and therapeutic recombinant DNA-derived
products licensed under Section 351 of the Public Health Service
Act [1] and currently regulated by the Center of Drug Evaluation and
Research (CDER). These biological products are also regulated as drugs
under the Federal Food, Drug, and Cosmetic Act [2]. The upstream
process in the manufacture of monoclonal antibodies and therapeutic
recombinant proteins typically involves cell expansion, cell culture, and
recovery steps. The downstream process involves multiple purifcation
steps. The purifed protein is ultrafltered/diafltered with formulation
bufer to provide a formulated bulk drug substance. The formulated
bulk drug substance is sterile-fltered and flled to provide a fnal drug
product. Because of the consequences of microbial contamination on
product safety and quality, there is continued interest in understanding
the root causes of microbial contamination and controlling these risks
in biologic product manufacture. This article discusses some of the
bacterial contamination cases reported to the Agency or identifed
during pre-license/pre-approval inspections of biologic drug substance
manufacturers in the past two years. The cases highlight areas for
Innovation is in Our Genes
PyroGene

rFC Assay
The evolution of endotoxin detection.
n Endotoxin specifc, recombinant technology eliminates false-positive glucan reactions
n Predictable, reliable assay performance lot after lot
n Sustainable resource no animal utilization
n Endpoint fuorescent assay, comparable to other quantitative LAL methods
n 510(K) submissions have been approved by the FDA, using PyroGene rFC Assay as a fnal release test
Discover for yourself.
To evaluate the PyroGene Recombinant Factor C Assay, visit www.lonza.com/rfc.
Lonza Walkersville, Inc. 8830 Biggs Ford Road, Walkersville, MD 21793
800 654 4452 301 898 7025
PyroGene is a trademark of the Lonza Group or its afliates. 2010 Lonza Walkersville, Inc.
PyroGene_Ad_AmerPharma_rev_0111.indd 1 1/18/11 10:45 AM

52 |

|

January/February 2011
mICroBIology

improvement in risk management and the need for developing a robust


microbial control strategy for biologic products.
sources of microbial Contamination
Microorganisms are ubiquitous in nature. Microorganisms can adapt
and survive under a variety of conditions and can pose a signifcant
risk to biologic products. An understanding of the microbial entry
points and implementation of measures to prevent microbial
contamination is critical for manufacture of safe, pure and potent
biologic products. As shown in Figure 1, microorganisms can gain
entry into a production process stream from several sources: the
facility, equipment, process operations, raw materials, column resins,
flter membranes, water, process gases, and personnel. All sources
of microbial contamination should be considered when developing
a microbial control strategy and performing an investigation for a
microbial contamination deviation.
regulation and guidance
The minimum current good manufacturing practice (CGMP)
requirements for preparation of fnished human drug products
are described in 21CFR211 [3]. These include the use of suitable
protective apparel (21CFR211.28), appropriate facility design and
placement of equipment (21CFR211.42), equipment cleaning,
sterilization, and maintenance (21CFR211.67), and production and
process controls (21CFR211.100). All these preventive measures
and precautions are implemented to protect product and prevent
contamination. Therapeutic recombinant products and monoclonal
antibodies are also subject to applicable regulation in 21CFR parts
600-610 [4]. The guidance on CGMP for active pharmaceutical
ingredients, Q7A, provides general CGMP guidance for biologic
drug substance manufactured by cell culture or fermentation under
section XVIII [5]. Additional guidance documents cover prevention or
control of adventitious agents in cell-derived biologic products and
address the quality concerns originating from cell substrates used for
manufacture of these products. These documents include (a) Points
to Consider in the Manufacture and Testing of Monoclonal Antibody
Products for Human Use published in February 1997, (b) Guidance on
Viral Safety Evaluation of Biotechnology Products Derived From Cell
Lines of Human or Animal Origin (Q5A), (c) Guidance on Quality of
Biotechnological/Biological Products: Derivation and Characterization
of Cell Substrates Used for Production of Biotechnological/Biological
Products (Q5D), and (d) Guidance on Specifcations: Test Procedures
and Acceptance Criteria for Biotechnological/Biological Products
(Q6B) [6,7,8,9]. The Q6B guidance states that contaminants should be
strictly avoided and/or suitably controlled with appropriate in-process
acceptance criteria or action limits for the drug substance or drug
product to meet specifcations. The 1994 FDA Guidance for Industry
on the Submission Documentation for Sterilization Process Validation
in Applications for Human and Veterinary Drug Products provides
guidance on sterilization process validation for fnal drug product
[10]. The 2004 FDA Guidance for Industry on Sterile Drug Products
Produced by Aseptic Processing Current Good Manufacturing
Practice provides guidance on personnel qualifcations, clean
room design, process design, and aseptic processing of fnal drug
products [11]. These regulations and guidance documents provide
the backbone for the development of an appropriate microbial
contamination control strategy.
Elements of a microbial Control strategy
A microbial control strategy should be developed once a
comprehensive risk assessment has been performed for all possible
microbial entry points into the manufacturing process. This requires
a good understanding of the manufacturing process and product
attributes. In general, the design of the facilities should allow for
proper operations and prevention of contamination. The fow of
personnel, material and waste should be from clean to dirty areas and
critical upstream open operations liable to microbial contamination
should be performed in designated biosafety hoods or areas with ISO
5 classifcation. Depending on the risks to the process, areas should
be appropriately segregated. Segregation of pre-viral and post-viral
clearance steps in processes using mammalian host cells is important
to prevent cross-contamination of process intermediates and the
facility. Segregation of areas, appropriate changeover procedures,
and other procedural controls should be in place to prevent cross-
contamination in a multi-product facility. Environmental monitoring
of manufacturing areas should be performed routinely at appropriate
intervals. Process gases and water should be tested and monitored to
ensure adequate microbial control. The design of equipment (single-
use disposable versus multi-use), validated cleaning and sterilization
processes along with a comprehensive preventative maintenance
plan are critical components of the microbial control strategy.
Microbial control for the lifetime use of membranes and resins should
be demonstrated. In addition, it is critical to identify and establish
processing steps that decrease bioburden and bacterial endotoxin
levels as the process intermediates are processed through sequential
purifcation steps. Bioburden reducing flters should be used at
Figure 1: Sources of microbial contamination.
124 Bernard E. Saint Jean Drive, East Falmouth, MA 02536 USA
tel: 888.395.2221 fax: 508.540.8680 custservice@acciusa.com www.acciusa.com
Run Both Chromogeni c AND
Turbi di metri c Assays i n
The Same Ki neti c Tube Reader
F L E X I B I L I T Y

54 |

|

January/February 2011
mICroBIology

critical steps in the process. This is critical for bufer solutions and in
process intermediates conducive to microbial growth. Minimizing
the number of open operations reduces the risk to product from
external (personnel and environmental) microbial contamination
sources. Biologic products are usually rich in carbon sources that
favor microbial growth. Hold conditions (time, temperature) for
a process should be validated to control and prevent potential
microbial growth. Bioburden and endotoxin alert and action limits
should be set for process steps based on process capability. Raw
materials should be screened for microbial quality and should be
handled and stored in a manner to prevent contamination and cross-
contamination. Personnel are important contributors to microbial
contaminations. Appropriate gowning should be implemented to
prevent contamination. All personnel performing open operations
should be trained adequately and evaluated periodically in
such operations.
Case studies
In the last two years, several contamination events were reported to
the Agency. They included viral or bacterial contamination of upstream
cell culture or fermentation processes. Viral contamination events were
extensively covered in the recent 2010 PDA/FDA Adventitious Viruses
in Biologics: Detection and Mitigation Strategies Workshop. Only
bacterial contaminations are discussed in this article. One case involved
contamination of a fermentor used in the manufacture of a protein
product secreted by a bacterial host. The contaminant was identifed
as Bacillus cereus (a Gram positive spore forming rod). A second case
involved the contamination of a fermentor used in the manufacture
of a recombinant protein by Paenibacillus curdlanolyticus (a Gram
variable spore-forming rod). A systematic approach was used during
the investigations to identify the root cause of the contamination and
included several media simulations to aid in identifying the point of
entry into the fermentor. In addition, the investigations involved the
manufacture of engineering batches. After a lengthy investigation
in both case studies, problems with the sampling devices, addition
valves, incorrectly ftted components, missing O-rings, incorrect
installation and deformation of an air flter after sterilization, and/or
inadequate slope of a condensate line were identifed. Immediate
corrective actions included the replacement of valve diaphragms
in fermentor addition ports, replacement of a membrane valve in
the sampling device, and replacement of O-rings on the measuring
probes. Enhancements were also made to the sterilization processes of
fermentor and associated transfer lines. A preventative maintenance
plan was developed for all fermentor valves. All valves were tagged
using a detailed checklist to ensure correct installation. All SOPs were
updated and employees were trained on the revised versions. The
investigations and corrective actions addressed all possible causes of
contamination as an unequivocal root cause could not be assigned.
In most cases, it is very difcult to identify
a defnitive assignable cause. It is highly
recommended that a systematic approach be
followed to determine the root cause. Media
simulations help in demonstrating that sterility
of the fermentor is not compromised. Recent
microbial contamination events at several
manufacturing facilities point to breaches
in the sterile boundary caused by damaged
vent flters, damaged O-rings, diaphragms,
and elastomers, and improperly sloped
condensate lines.
When bacterial hosts are used, microscopic
examinations of the fermentation culture for
contamination is difcult. A culture purity test
should be perfomed using appropriate media
and culture conditions. It is crucial to have a
comprehensive preventative maintenance plan
for fermentor and tank agitators, probes, gaskets,
O-rings, valves, and flters. The design of piping
and valves should prevent steam condensate
from collecting and leading to contamination
by back-fow. After periods of shutdown or
maintenance, it is important to perform media
simulations on sterile equipment that has
remained idle for a period of time. Procedural
details on assembly and set-up of fermentors/
bioreactors should be clear and very detailed.
Training in this area can reduce inadvertent

January/February 2011 |

| 55

mICroBIology
leaks and contamination of the systems. Continuous assessments of
change control, work orders, and other process improvements should
be conducted to ensure that the microbial control strategy is not
impacted. Of note in both cases, the contaminating microorganism was
a facultative anaerobic Gram positive spore-forming rod. Risk mitigation
strategies based on microbial environmental fora should be considered.
The areas for improvement identifed in the case studies were in
preventative maintenance plans for all fermentor valves including valves
on sampling devices and in the documentation
for correct assembly of components.
Two cases of microbial contamination of the
downstream process were identifed during
pre-approval/pre-license inspections of drug
substance manufacturing facilities. Bioburden
deviations were observed in several batches at
the ultra-fltration/diafltration (UF/DF) step. The
contaminants identifed were Sphingomonas
species, Stenotrophomonas maltophilia,
Ralstonia pickettii, and Staphylococcus species
suggesting probable water and human sources
of contamination. Presence of repeated high
bioburden counts in several batches suggested
development of bioflm and inadequate
contamination control procedures for the
UF/DF steps. After extensive investigations,
several corrective actions were implemented
in terms of cleaning, storage and re-use of UF/
DF systems, sterilization/sanitization of bufer
tanks, assessment of the water for injection
(WFI) system and transfer lines, introduction
of in-process bioburden reducing flters (in
cases where there were no flters before the
UF/DF steps), validation of hold times and
storage conditions of process intermediates
and revisions to bioburden limits based on
process capability. Demonstration of microbial
control over the lifetime use of membranes and
validation of in-process hold times are essential
for ensuring the consistent quality of biologic
products. All WFI piping locations with stagnant
water should be assessed and eliminated.
Microbial trend reports for water systems should
be reviewed regularly.
The investigations of microbial contaminations
are challenging due to the ubiquitous nature
of the microorganisms, multiple points of
microbial entry, growth promoting properties
of biological process streams, limitations of
sampling and detection methods, and the time
and resources involved in performing complex
investigations. All microbial entry points should
be systematically evaluated. For fermentor
contaminations, seed fermentors and associated
additions and transfer lines should be included
in the investigations. A hazard analysis and critical control point
assessment for bioburden control throughout the manufacturing
process is useful for the design of a microbial control strategy and
the performance of a systematic investigation. In addition, failure
data should be tracked to gain a better understanding of root causes.
The information should be used to continuously evaluate risks and
implement process and/or equipment improvements to mitigate and
prevent microbial contaminations.
Accelerate and automate
your microbial testing.
The Growth
Direct
TM
System
Rapid Automated
Compendial Test
Non-Destructive
Replaces human eye with digital imaging
to deliver rapid microbial enumeration:
Automated incubation, enumeration, reporting
& LIMS data transfer saves labor & improves compliance
Applications & throughput cover your routine
QC testing needs
Streamlined validation compatible with
USP methods and regulatory guidelines
Non-destructive test allows faster investigations
Rapid Micro Biosystems, Inc.
Bedford, MA 01730
Phone: 781-271-1444
www.rapidmicrobio.com
Save time. Save money. Increase productivity.

56 |

|

January/February 2011
mICroBIology

Conclusions
Microbial contamination is a risk to biologic product quality and
safety. The cost of inadequate microbial control in biologic product
manufacture is enormous as facilities or bioreactor production
trains may have to be shut down for lengthy periods of time in
order to conduct investigations and identify the root cause to
prevent reoccurrence. The recent cases of bacterial contamination
of biologic products suggest that preventative maintenance plans
for fermentor and associated valves, types of materials used for
diaphragms and O-rings, and understanding of microbial control at
certain process steps need further attention. Contamination control
requires an understanding of the microbial entry points and risks to
the process as well as the microbial growth potential of the product,
media and bufer solutions. Microbial contamination control requires
appropriate design of facility and equipment, validated cleaning and
sterilization cycles for equipment, detailed and robust preventative
maintenance plans for equipment, measures to reduce bioburden
and bacterial endotoxins at appropriate steps in the process, and
routine monitoring of these process steps for bioburden and
endotoxin with defned alert and action limits. A contamination
remediation plan should be established. Such a plan is benefcial for
meeting CGMP and has the advantage of reducing facility downtime.
Investigations should be comprehensive and include assessment
of all microbial entry points. Corrective actions should address all
possible identifed causes in the absence of a known assignable
root cause. The information gathered during these investigations
should feed into the overall risk management plan. The quality risk
management plan should be integrated into the quality system and
allow for continuous improvement.
references
1. Public Health Service Act, Biological Products; as amended
2. Federal Food Drug and Cosmetic Act; as amended.
3. FDA, Current Good Manufacturing Practices for Finished
Pharmaceuticals, 21 CFR part 211.
4. FDA, Biologics, 21 CFR parts 600-610.
5. U.S. Department of Health and Human Services, Food and Drug
Administration. Guidance for Industry: Q7A Good Manufacturing Practice
Guidance for Active Pharmaceutical Ingredients. Rockville, MD; 2001.
6. U.S. Department of Health and Human Services, Food and Drug
Administration. Centre for Biologics Evaluation and Research. Points
to Consider in the Manufacture and Testing of Monoclonal Antibody
Products for Human Use. February 1997.
7. U.S. Department of Health and Human Services, Food and Drug
Administration. Guidance for Industry: Q5A Viral Safety Evaluation
of Biotechnology Products Derived From Cell Lines of Human or
Animal Origin. Rockville, MD; 1998.
8. U.S. Department of Health and Human Services, Food and Drug
Administration. Guidance for Industry: Q5D Guidance on Quality of
Biotechnological/Biological Products: Derivation and Characterization
of Cell Substrates Used for Production of Biotechnological/Biological
Products. Rockville, MD; 1998. Federal Register Vol. 63, No. 182, 1998.
9. U.S. Department of Health and Human Services, Food and Drug
Administration. Guidance for Industry: Q6B Specifications: Test
Procedures and Acceptance Criteria for Biotechnological/Biological
Products, FDA, 1999.
10. U.S. Department of Health and Human Services, Food and
Drug Administration. Guidance for Industry for the Submission
Documentation for Sterilization Process Validation in Applications for
Human and Veterinary Drug Products. Rockville, MD; 1994.
11. U.S. Department of Health and Human Services, Food and Drug
Administration. Guidance for Industry: Sterile Drug Products
Produced by Aseptic Processing Current Good Manufacturing
Practice. Rockville, MD; 2004.
Biography
Kalavati Suvarna, Ph.D. is a Microbiologist with the Biotech
Manufacturing Team in the Division of Manufacturing and Product
Quality in the Ofce of Compliance, CDER, FDA. She has over nine years of
experience as a microbiology reviewer at the FDA. Kalavati holds a Ph.D.
in Biological Sciences from Northern Illinois University. Prior to joining the
Agency, she worked in an academic and pharmaceutical setting.
Anastasia G. Lolas is a Microbiologist with the Biotech Manufacturing
Team in the Division of Manufacturing and Product Quality in the Ofce
of Compliance, CDER, FDA. She has over 5 years of experience as a
microbiology reviewer of drug applications at the FDA. Anastasia holds
a B.S. in Biology from Virginia Polytechnic Institute and State University
and a M.S. in Food Science from the University of Illinois at Urbana-
Champaign.
Patricia F. Hughes, Ph.D. is the Team Leader in the Biotech Manufacturing
Team in the Division of Manufacturing and Product Quality in the Ofce
of Compliance in CDER, FDA. She has over twenty years experience in the
Pharmaceutical/Biotech industry in fermentation & cell culture process
development and manufacturing. In addition, she has over twelve years
of experience as a microbiology reviewer at the FDA, in CDER and CBER.
Patricia holds a Ph.D. in Microbiology from Georgetown University.
Richard Friedman is the Director of the Division of Manufacturing &
Product Quality in the Center for Drug Evaluation and Research (CDER),
Ofce of Compliance. In this position, he directs the interpretation and
development of CGMP policy, review of inspectional recommendations
and determination of manufacturing site acceptability. He has been
employed by FDA since 1990, including prior positions as New Jersey
District Drug Specialist, CDER Senior Compliance Ofcer and Team Leader
of Guidance and Policy. Mr. Friedman has authored several publications
on topics including sterile drugs and quality management systems, and
was awarded The George M. Sykes Award by the Parenteral Society for
outstanding journal paper for the year 2005. Mr. Friedman is also an
adjunct faculty member of Temple University School of Pharmacy in their
QA/RA graduate program. Prior to joining FDA, Mr. Friedman worked
in the toxicology research division of an innovator pharmaceutical
company. Mr. Friedman received his B.S. in Biology with honors from
Montclair State University in 1989 and his M.S. in Microbiology from
Georgetown University School of Medicine in May, 2001.
Do you live an InterContinental life?
Whether youre in Baltimore for business or pleasure, a stay at the four-star InterContinental Harbor Court Baltimore
Hotel is guaranteed to leave you with an unparalleled experience of the city. With our award-winning restaurants,
impeccably appointed rooms and enticing amenities, the Harbor Court Baltimore reveals the beauty of Baltimore to
guests at every turn. And with our central location in the heart of the historic Inner Harbor, within walking distance to
the downtown business district and countless popular attractions, theres no easier way to access everything you require
to make your visit a success. Get the most from your Baltimore experience, the InterContinental way.
A STEP ABOVE THE
REST, I N THE HEART
OF BALTI MORES
I NNER HARBOR.
Call 1.800.424.6835 or visit
www.intercontinental.com/baltimore
2007 InterContinental Hotels Group. All rights reserved.
.

58 |

|

January/February 2011
sInglE-usE

Disposable manufacturing platforms are becoming increasingly


popular for therapeutic monoclonal antibody (MAb) manufacture,
whether as fully disposable platforms or for incorporating single
use solutions in to largely fxed (i.e., stainless steel-based, re-usable)
manufacturing platforms. While large pharmaceutical companies
have largely adopted a platform approach to development and
commercialisation of therapeutic monoclonal antibodies, or employ
single use solutions for certain unit operations (e.g., seed train
expansion), disposable platforms may be more common among small
to medium sized, or more recently established, companies. This may
have implications for partnerships between small-to-medium and
large pharma companies seeking to co-develop and commercialise
biopharmaceuticals.
We describe here the transfer, manufacture of clinical trial drug
substance lots, and late stage development in advance of process
validation, of a fully disposable MAb manufacturing process in an
existing small molecule API clean room facility. Four MAb drug
substance lots were manufactured within 14 months of facility
selection decision. The overall transfer process, and specifc challenges
arising from implementing disposable-based biopharmaceutical
manufacturing in an existing small molecule API facility are discussed.
This approach may become more relevant as companies assess existing
facilities in the context of a changing business environment (moving
in to biopharmaceutical manufacturing, strategic responsiveness to
partnering opportunities). The case described here represents an
alternate route to biopharmaceutical manufacturing, creating options
for leveraging existing facilities, as opposed to constructing dedicated
biopharmaceutical manufacturing space.
The MAb molecule discussed here is a therapeutic IgG1, manufactured
by a GS-CHO-based (fed-batch), two column process. The molecule
was co-developed between Eli Lilly & Co. and MacroGenics, Inc.
An overview of the process is shown in Figure 1. The process is
based largely on single use components, with the afnity capture
chromatography column and purifcation skids being re-used (i.e.,
cleaned between uses).
As shown in Figure 2, Eli Lilly began co-developing the MAb for
commercialisation in 2008. At this time, MacroGenics had already
generated material for Phase III supply, and additional Phase III
material was to be generated in Lillys Kinsale facility, which would
also be the site for process validation, launch, and commercial supply.
Lilly Kinsale has a 30+ year history in small molecule API market supply
(post-launch) for both parenterals and non-parenterals and was in
Brian Mullan, Ph.D., Kristi Huntington, Aidan
Collins, and Marie Murphy, Ph.D.
Eli Lilly & Co.
email: mullan_brian@lilly.com
Transfer, Implementation
and Late Stage
Development of an
End-To-End Single-Use
Process for Monoclonal
Antibody Manufacture
w: www.pall.com/economics
e: biopharm@pall.com
For those who appreciate
a high degree of
selectivity
Defining Process Economics
2011 Pall Corporation. Pall, , HyperCel, and Mustang are trademarks of
Pall Corporation. indicates a trademark registered in the USA. GN10.3516
Unique chromatography
selectivities deliver economy
and performance at every scale
Palls chromatography chemistries feature
differentiated selectivities for ion exchange
and mixed-mode chromatography that
help achieve protein purification goals
and decrease process costs.
u Increase productivity: Q and S HyperCel

sorbents provide high dynamic binding


capacity at 2 to 5 times the flow rates of
conventional ion exchangers.
u Reduce process steps: HyperCel mixed-
mode sorbents can eliminate unit operation
steps from your process.
u Decrease salt concentration: MEP, HEA
and PPA HyperCel sorbents are an alternative
to HIC that reduces the amount of salt
required for protein capture.
u Decrease buffer usage: Mustang

ion
exchange membrane capsules significantly
reduce buffer consumption during
contaminant removal.
u Simplify scale up: Flexible designs allow
consistent performance from lab to process
scale and reduce revalidation requirements.
Visit Pall online to get the complete story.
10-3516_Chrom-AmericanPharmRev-Fullpg_AD:layout 17/01/2011 12:34 Page 1

60 |

|

January/February 2011
sInglE-usE

the process of developing launch and commercial supply capability


for biopharmaceuticals at this time (2007; construction of a re-usable
platform-based facility in Kinsale). The agreement and project
timing with MacroGenics was in advance of scheduled availability of
this new facility (2010). Additionally, this opportunity enabled Lilly
Kinsale to begin to build capabilities in the areas required to support
biopharmaceutical manufacturing, using a smaller scale and fexible
platform, in advance of its larger scale platform facility coming online
in 2010.
Figure 1. Process fow for MAb manufacturing process
Figure 2. Overall timeline for process transfer, facility readiness
and schedule of Phase III campaigns leading to process validation
in Q4 2010. The red diamond indicates timing of facility selection
decision. Activities at Lillys Kinsale facility are shown in blue
boxes, activities at Lilly Bioprocess R&D, Indianapolis, are shown
in orange boxes. Drug substance technical program (green
boxes) was jointly executed between Kinsale and BR&D. DS,
drug substance; BR&D, Bioprocess R&D; CT, clinical trial; ENG,
Engineering batches; PS, primary stability; PV, process validation;
SM, small molecule.
GE Healthcare
Life Sciences
Part of the ReadyToProcess
TM
platform, WAVE Bioreactor is a fast and effcient
system for innoculum propa gation and cell culture. Scale-up is simple as no
passaging is needed just add fresh culture media to the Cellbag

bioreactor.
WAVE Bioreactor reduces time-consuming routines, increases fexibility,
and ensures the integrity of your operations.
ReadyToProcess, just plug in & youre ready to go.
Find out more at www.gelifesciences.com/readytoprocess
Plug & play with WAVE Bioreactor

and accelerate your seed train


Cellbag, ReadyToProcess, and WAVE Bioreactor are trademarks
of GE Healthcare companies.
2010 General Electric Compnay All rights reserved.
GE Healthcare Bio-Sciences AB, Bjrkgatan 30, 75184 Uppsala, Sweden.
GE10-10. First publised November 2010.

January/February 2011 |

| 61

sInglE-usE
At a process development level, the process as transferred was appropriate
for Phase III supply, but required additional characterisation and
development to be ready for process validation. This work was initiated
in Eli Lillys Bioprocess R&D (BR&D) facility in 2008, with co-development a
joint efort between Lillys BR&D and Kinsale development units.
Numerous considerations were assessed to transfer a large molecule
manufacturing process in to a clean room facility previously used
for a small molecule parenteral API (Table 1). Among these, key
considerations to meet the overall timeline were supply chain and
analytical testing. For the frst (Engineering / Clinical Trial supply)
Phase III campaign in Kinsale, the supply chain was managed by BR&D,
Indianapolis, with raw materials and consumables being transferred
to Kinsale from Indianapolis via inter-warehouse transfer (followed by
receipt verifcation and GMP storage in Kinsale). All analytical testing
for the MAb molecule (batch release, in process) was performed by
MacroGenics, Lilly BR&D, or was outsourced (e.g., for adventitious
agent testing). The exception to this was microbiological testing
(bioburden, endotoxin), for which methods were transferred to and/or
qualifed in Kinsale to support direct testing at the Kinsale site, and raw
materials release testing, which was also performed at Kinsale.
At a facility level, the overall approach was to establish a GMP system
for campaign-based use of an existing small molecule API parenteral
facility on the Lilly Kinsale site. This facility consisted of in-built
equipment (e.g., glove boxes) in numerous, large (ca. 40m2 per clean
room) ISO 7 or 8 classifed clean areas. During small molecule API
manufacture intermediates are housed and moved in closed transport
containers between unit operations. The facility contained a parts
washer and an autoclave, and dedicated processing areas for diferent
areas of process support (e.g., non-clean and clean parts areas), and for
diferent unit operations of the API manufacturing process.
For biopharmaceutical manufacturing, areas in the facility were
assigned as follows:
Small, simple, precise pumps
The compact 120
peristaltic pump for
biopharm, science and
research saves bench
space and improves yield.
Exceptional 2000:1 speed
range oers precise ows
to 190ml/min.
N
E
W
Watson-Marlow Flexicon Tubing
wmpg.com
800-282-8823
BioPharmaceutical Division
S
ee us
at Interphex
booth #2807
120-WM-APR7x4.875_Layout 1 2/9/11 9:15 AM Page 1
Table 1. Facility ft and other considerations for transfer of a MAb
manufacturing process in to a small molecule parenteral API
manufacturing facility. Items are shown in alphabetical order.
DCS, Distributed control system; HVAC, Heating ventilation and air
conditioning; SME, subject matter expert;
Alarm management (accessibility to existing system, ability to route alarms to supervisory
areas)
Autoclave, parts washer availability
Cold storage (2-8oC, -20oc, -80oC)
DCS points / accessibility to plant historian system
Operations readiness and training
Personnel and equipment fows
Potential cross-contamination with existing products (multi-product management)
Quality systems
Resources, capabilities (SMEs, technical team, Analytical [in-house or out-sourced])
Utilities (HVAC, number of power points, process gases)
Viral boundary (pre-, post-)
Warehousing space
Waste management (including biohazardous waste) and disposal

62 |

|

January/February 2011
sInglE-usE

Bufer preparation* (ISO 8)


Cell culture media and feed preparation* (ISO 8)
Vial thaw and seed train expansion (ISO 7 with ISO 5
biological safety cabinet added)
Production Bioreactor and Primary Recovery (ISO 8)
Initial purifcation: Afnity capture to Nanofltration (ISO 8)
Final purifcation: UFDF to fnal formulation and fll (ISO 8)
* weigh and dispense, dissolution, in process checks.
A pre-viral/post-viral boundary, controlled by GMPs, was designated
between the last two areas.
As the existing facility contained adequate open spaces, supporting
manufacture of the small molecule parenteral API largely
through the use of fxed equipment, the change-overs to support
biopharmaceutical manufacturing (Figure 2) were based largely on a
wheel-in, wheel-out approach, with some facility modifcations, as
shown in Figure 3.
specifc Challenges:
supply Chain and
Analytical testing
The small molecule process supported by
the Kinsale clean room facility had about fve
raw materials and 15 consumables. The MAb
manufacturing process had about 20 raw
materials and 200 consumables, ranging in
size from disposable bioreactor bags (fully
packaged) down to pipette tips. Logistic
and supplier quality aspects of supply chain
were supported by Lilly Indianapolis (with
inter-warehouse transfers to Kinsale) as
establishment of a supply chain de novo for
Lilly Kinsales frst biopharmaceutical would
not have been possible in the agreed project
timeline. Even with this support, physical
storage and management of this amount of raw
materials and consumables was challenging,
with the MAb process efectively taking over
an entire warehouse, including a walk in 2
8
o
C area, and required installation of -20
o
C and
-80
o
C storage.
The Lilly Kinsale site had a well established
Analytical group supporting small molecule
manufacturing, including microbiology
testing and environmental monitoring (EM).
Transfer and validation of the ca.15 methods
supporting release testing of the MAb process
would not have been possible in the agreed
timeline as the majority of methods required
new equipment (e.g., for CE-LIF, CEX, CE-SDS
methods) and expertise. All testing, excepting
EM and microbiological testing, and raw
material release testing, was performed by
MacroGenics, Lilly BR&D, or was out-sourced
(for adventitious agent testing and bioassay).
the new math of
rapid detection
The Cell Culture Rapid Methods Program from Life Technologies
lets you detect the three most prevalent threats to CHO cell culture
manufacturing, typically in less than 4 hours, enabling nearreal-time
monitoring from start to fnish.
1 sample prep + 1 instrument platform =
detection of Mycoplasma, Vesivirus, and MMV
DNA | RNA | PROTEIN | CELL CULTURE | INSTRUMENTS
Go to www.appliedbiosystems.com/seq
FOR RESEARCH USE ONLY. NOT INTENDED FOR ANY ANIMAL OR HUMAN THERAPEUTIC OR DIAGNOSTIC USE. 2011 Life Technologies
Corporation. All rights reserved. The trademarks mentioned herein are the property of Life Technologies Corporation or their respective
owners. CO17317 0210
CO17317 CCRM Ad 1/2page Resize for American Pharma.indd 1 2/10/11 5:05 PM
January/February 2011 |

| 63

sInglE-usE
specifc Challenges: Disposable Processes
No specifc challenges were encountered during implementation and execution of unit
operations where disposable technologies are more routinely applied, e.g., seed train expansion,
primary recovery. Issues more so arose with the production bioreactor, some protein purifcation
unit operations, and generic concerns related to use of disposable manufacturing systems.
At a generic level, sourcing and availability of standard tubing assemblies (extender pieces,
pump tubing assemblies) aided process fow during manufacturing of the initial (non-GMP)
engineering lots, enabling set-up of equipment trains to be optimised (process ft and connection
harmonisation). Of-the-shelf tubing solutions are not widely available from vendors, and
custom solutions from most vendors can take 4 16 weeks for delivery, by the time vendor
set-up, assembly design, fabrication, -irradiation are complete. Additionally, key technical
details for consumables can become buried in the large amount of items (in our case 200+) that
need to be simultaneously set up and managed. An example in point is a key flter for protein
purifcation, that at the original 1.2m
2
confguration was supplied as -irradiated by the vendor,
but at 0.6m
2
confguration (as used at Lilly Kinsale) was not supplied as -irradiated. This aspect
was not initially noticed during materials set-up. In this instance, an additional -irradiated
0.2/0.45um guard flter was placed in line (downstream) of the 0.6m
2
flter to provide additional
aseptic assurance.
For the production bioreactor, the process as transferred comprised a disposable cell bag from one
vendor installed in to a bioreactor system from another. Initial interest in moving to in-line probes (for
pH and temperature) was not supported by our experience with these probes (numerous bag ruptures),
and a customised probe was qualifed for temperature control, with pH being monitored of-line via a
blood gas analyser.
For the purifcation unit operations, the process as transferred contained some manual mixing
steps, which were all replaced by non-manual mixing systems (rocking platforms, bulk mixing
systems). This necessitated mixing development and optimisation of control parameters in
many instances.
Lastly, the platform was further developed and optimised by developing customised single use
solutions for certain issues encountered during process transfer or which were required prior to
process validation. These included:
Design of a integral flter + bag solution (and sampling port assembly) for intermediate
hold of clarifed harvest;
Design of a bulk mixing system for pH adjustment of a purifcation intermediate;
Integration of individual components required for fnal fll and fltration of formulated
drug substance in to a single -irradiated assembly.
single use systems
Contact us by calling
800-724-4158
As a part of Advanced Scientics
Single Use Systems, the company
offers a multitude of solutions for
single use bags. With sizes ranging
from 50 mL to 3,500 L, Advanced
Scientics can produce single use
bags of nearly any conguration.
These bags are currently utilized
throughout the industry and
throughout the manufacturing
process.
In addition to specializing in custom
solutions, we offer a large variety of
catalog products.
Advanced Scientics is an FDA registered,
ISO 13485:2003 certied manufacturer.
w
w
w
.
a
d
v
a
n
c
e
d
s
c
i
e
n
t
i


c
s
.
c
o
m
Figure 3. Adaptation of an ISO 8 classifed area in a facility used for small molecule parenteral
API manufacturing. [Left Panel] Open space is shown, containing glycol loops. [Right panel]
200L disposable bioreactor systems in place in same area. A process gas (air, O2, CO2)
manifold has been added to the rear wall.

64 |

|

January/February 2011
sInglE-usE

In many of these instances opportunities were taken to reduce both


the complexity of operational set-ups and aseptic risk by incorporating
multiple components in to one disposable solution that was provided
as a single -irradiated assembly. The downside to this is that such
customised solutions reduce supply chain fexibility, commonly
taking 12 16weeks for order fulflment and 2-4 weeks to complete
design. Inclusion of these solutions should ideally represent a balance
between the beneft provided by the customised solution and supply
chain fexibility.
Product and Process Comparability
Following completion of the frst MAb drug substance manufacturing
campaign in Q1 2009, process and product comparability between
MacroGenics, Lilly BR&D (where numerous at scale developmental
runs had also been performed), and the batches manufactured at Lilly
Kinsale was assessed. Both the process and the product manufactured
at Kinsale were deemed to be parametrically and analytically
comparable to batches manufactured by MacroGenics and Lilly
BR&D. The batches manufactured in Lilly Kinsale incorporated certain
process changes (some described above), and inclusion of modifed
solution mixing parameters (following mixing studies and installation
of standardised mixing systems).
The Lilly Kinsale site and API/MAb facility was inspected by the Irish
Medicines Board in June 2009, with the agency granting a clinical
trial manufacturing (IMPD) licence for the MAb process following a
successful inspection outcome.
Concluding remarks
This article describes transfer, implementation and manufacture
of Phase III drug substance material for a monoclonal antibody
manufacturing process in a facility that was also employed for
manufacture of a small molecule parenteral API. The MAb process
is a largely fully disposable process, excepting afnity capture
chromatography column and purifcation skids, which are cleaned
and re-used.
Numerous considerations for transfer of single use MAb processes
in to existing facilities were assessed, with the most prominent
considerations being supply chain and analytical testing.
The case described here shows that adaptation of existing clean
room facilities, coupled with the fexibility inherent in single-use
MAb processes, is both feasible and can have successful outcomes in
terms of agency inspections. This approach represents an alternate
route to biopharmaceutical manufacturing, creating options for
leveraging existing facilities, as opposed to constructing dedicated
biopharmaceutical manufacturing space.
Acknowledgments
The authors would like to acknowledge their many colleagues in Lilly
Kinsale (Operations, Quality, Procurement & Warehousing, MS&T,
and Analytical), in Lilly Bioprocess R&D, and at MacroGenics who
contributed to the work described in this article.
Author Biographies
Brian Mullan is a Scientist/Tech Transfer Lead,
Manufacturing Science and Technology (MS&T), with
Eli Lilly & Co, Kinsale, Cork, Ireland. Since joining Lilly
he has been responsible for late stage development and
transfer of biopharmaceutical manufacturing processes
to launch sites. He previously worked for Centocor (Johnson & Johnson) in
Cork, Ireland, and with Sanof Aventis in Paris, Toulouse, and New Jersey.
Brian holds a BSc in Biochemistry from University College Galway, Ireland,
a PhD in Viral Genetics/Cell Biology from University College Cork, Ireland.
Aidan Collins is a Scientist, MS&T, with Eli Lilly & Co,
Kinsale, Cork, Ireland. At Lilly he has been responsible for
implementation of new biopharmaceutical processes
(protein purifcation) for pipeline and inlicensed
products. He has previously worked for BioUetikon,
Baxter S.A., Pfzer, and Amgen prior to joining Lilly in 2007. He holds a
BSc in Biochemistry and Molecular Biology, and a Masters in Quality
Assurance from Dublin Institute of Technology, Ireland.
Kristi Huntington is an MS&T Consultant with Eli Lilly
& Co, Indianapolis, IN, USA. At Lilly she is currently
responsible for new product introduction (NPI) for drug
substance manufacturing. She has previously held
roles as NPI lead and MS&T Manager at Eli Lilly & Co,
Kinsale, Cork, Ireland, and Technical Services Representative, Eli Lilly &
Co, Carolina, Puerto Rico, and Indianapolis, IN, since joining Lilly in 2000.
She holds a BSc in Chemistry from Hiram College, Ohio, and an MSc in
Chemistry from The Ohio State University.
Marie Murphy is a Microbiology and Virology Lead
with Eli Lilly & Co, Kinsale, Cork, Ireland. At Lilly she is
responsible for the delivery and implementation of
product protection strategies including viral safety for
biopharmaceutical manufacturing. She has previously
worked for EiRx Therapeutics, Cork, Ireland, and for Schering Plough, Cork,
Ireland. Marie holds a BSc in Microbiology from University College Cork,
Ireland, and a PhD in Virology from Trinity College, Dublin, Ireland.
join over 1000 of
your colleagues
Discovery
Phage & Yeast Display
Engineering Antibodies
Antibody Optimization
expression
Diffcult to Express Proteins
Optimizing Protein Expression
Purifying Antibodies
analytical
Characterization of Biotherapeutics
Protein Aggregation and Stability
Immunogenicity
antiboDies
Antibodies for Cancer Therapy
Bispecifc Antibodies
Antibody-Drug Conjugates
Organized by Cambridge Healthtech Institute
250 First Avenue, Ste 300, Needham MA, 02494 PEGSummit.com
the essential protein engineering summit
Experience the future of biologics.
sheraton boston hotel | boston, ma
keynote speakers
Raimund Dutzler Ph.D.,
Professor, Biochemistry,
University of Zurich
Allan Bradley Ph.D.,
FRS, Director Emeritus,
Wellcome Trust Sanger
Institute
Wayne Coco Ph.D.,
Biologics Research, Bayer
Schering Pharma AG
Davinder Gill Ph.D.,
Vice President and Head,
Global Biologics
Technologies, Pfzer, Inc.
James D. Marks M.D., Ph.D.,
Professor and Chief of Anesthesia,
San Francisco General Hospital;
Vice Chairman, Anesthesia and
Perioperative Care, UCSF
Lorenz M. Mayr Ph.D.,
Executive Director, Unit Head
Biology, Protease Platform,
Novartis Pharma AG
Janice Reichert Ph.D., Research
Assistant Professor, Tufts Center
for the Study of Drug Develop-
ment, Tufts University School of
Medicine
Roger A. Sabbadini Ph.D.,
Founder, Vice President &
CSO, Lpath, Inc.; Professor
Emeritus, Biology, San Diego
State University
William R. Strohl Ph.D., Vice
President, Biologics Research,
Biotechnology Center of
Excellence, Centocor
Douglas Cecchini Ph.D.,
Director, Technical Development,
Biogen Idec, Inc.

66 |

|

January/February 2011
FormulAtIon DEvEloPmEnt

Introduction
It is frequently reported that the percentage of drug candidates that
are limited by poor solubility is increasing [1,2]. These poorly soluble
compounds typically require enabling formulations, and this trend
creates challenges for teams in discovery and development who must
drive in-vivo exposures high for animal toxicology studies and deliver
robust dosage forms for clinical evaluation. Many enabling technologies
are available for the formulator to consider, including lipids, cosolvents,
surfactants, nanoparticles, cyclodextrin complexes, amorphous solid
dispersions, and others. The suitability of the particular formulation
approach depends largely on the physicochemical properties of the
active pharmaceutical ingredient (API). Amorphous solid dispersions
(ASDs) are particularly attractive for many poorly soluble drug
candidates because these formulations ofer many of the advantages of
more conventional solid oral dosage forms but they also provide faster
dissolution rates and higher drug concentrations in the gastrointestinal
milieu [3]. Further, typical excipients utilized in production of ASDs are
commercially available and they have proven to be well tolerated in
vivo. We have successfully employed ASD technology to drive high
plasma exposures in toxicology studies as well as to deliver challenging
molecules in clinical studies. In this article we will discuss approaches
for preparing, screening, characterizing, and dosing ASDs in preclinical
and early development.
methods of Preparation
Rotary Evaporation
Rotary evaporation is a desirable method for preparation of ASDs
for early stage (pre-clinical efcacy/toxicology, Phase I) studies. This
approach is fast, material sparing, relatively inexpensive, and readily
available. Moreover, a wide range of batch sizes from mg to kg
quantities may be prepared with high yield. ASD preparation by rotary
evaporation is carried out by frst dissolving the API and formulation
components (polymers, surfactants) in a pharmaceutically acceptable
solvent. Typical solids load in the solvent is 5% to 25% by weight, and
this is generally dictated by API/polymer solubility. The solvent is
then removed in a rotary evaporator using heat (typically 40 to 80
o
C) and vacuum. Total time for solvent evaporation can range from
minutes to hours.
Brian E. Padden, Ph.D., Jonathan M. Miller, Ph.D.,
Timothy Robbins, Ph.D., Philip D. Zocharski,
Leena Prasad, Julie K. Spence,
& Justin LaFountaine
Abbott Laboratories, Abbott Park, IL
email: brian.padden@abbott.com.
Amorphous Solid
Dispersions as Enabling
Formulations for Discovery
and Early Development
improved
bioavailability.
softgel
solutions.
SOLVE WATER
SOLUBILITY
CHALLENGES
Improve the performance of
your BCS II/IV compounds
with our innovative softgel
delivery solutions.


2
0
1
1

C
a
t
a
l
e
n
t

P
h
a
r
m
a

S
o
l
u
t
i
o
n
s
.

A
l
l

r
i
g
h
t
s

r
e
s
e
r
v
e
d
.
DELIVER
MORE APIs
Bring your challenging
APIs to market with our
innovative, non-gelatin
Vegicaps capsules,
which can handle a
wider range of lls and
higher melting points.
SOLVE
PERMEABILITY
CHALLENGES
Increase the permeability
of your BCS III/IV
compounds with our unique
lm coating technology
and ll formulations.
Discover more solutions with Catalent.
Call: + 1 866 720 3148 Email: info@catalent.com Visit: www.catalent.com DEVELOPMENT DELIVERY SUPPLY
Catalent is your catalyst for better bioavailability. Our team of 1,000+ development and formulation
scientists have the talent and expertise to solve the most complex bioavailability and permeability
challenges to help you deliver more effective treatments. With over 75 years of experience and a wide
range of innovative drug delivery solutions, we can improve the performance of your products, from
discovery to market and beyond. Catalent. More products. Better treatments. Reliably supplied.

8759 Catalent Softgel APR 8.125x10.875.indd 1 1/20/11 4:18 PM

68 |

|

January/February 2011
FormulAtIon DEvEloPmEnt

Because of the relatively long evaporation time, the compound


must have adequate chemical stability in the solvent at elevated
temperatures. Longer evaporation times may lead to physical
instability, so care must be taken to avoid API crystallization during
solvent removal. This issue can be mitigated to a large extent through
optimization of the temperature, vacuum, rotation, and total solids
load. After removal of the solvent, the resulting ASD is isolated, dried,
and milled to the desired particle size. Secondary drying in a vacuum
oven or tray dryer is often employed to remove any residual solvent
that remains in the fnal ASD powder.
The polymers that may be employed for ASD preparation by rotary
evaporation are limited to those that can be easily isolated in high yield
after solvent removal (Table 1). Hydroxypropylmethyl cellulose (HPMC)
based polymers are typically not amenable to rotary evaporation, as
these polymers often result in a glassy flm that is difcult to isolate.
While preparation of ASDs by rotary evaporation is ideal for the early
stages (pre-clinical to Phase I) of drug development, it is not well suited
for later stage development, manufacturing, and commercialization.
The process is not readily scaleable beyond quantities on the order of
10 kg because solvent volumes become too large, leading to very long
and unrealistic evaporation times. Therefore, bridging to larger scale
spray drying or hot melt extrusion (HME) processes is required if the
drug candidate progresses into later stage development.
Spray Drying
Spray drying is another method that is commonly utilized for the
preparation of ASDs of poorly soluble compounds [4]. The method
is readily scaleable from gram-sized batches during discovery
and early development to kg and metric ton quantities during
later stage manufacturing and commercialization. The frst step
in the spray drying process is to prepare a feed solution of the
API and formulation components (polymers, surfactants) in a
pharmaceutically acceptable solvent. The total solids load in the
feed solution is typically 5% to 25% by weight, and this is generally
dictated by API/polymer solubility as well as viscosity of the solution.
The feed solution is then pumped into a spray nozzle along with
inert, hot (typically 60 to 100
o
C) drying gas where it is atomized
and sprayed into a drying chamber. The solvent quickly evaporates
during this process, leaving behind spray dried dispersion particles.
These particles are collected in a cyclone with attached baghouse
flter. Secondary drying in a vacuum oven or tray dryer is often
employed to remove any residual solvent that remains in the fnal
ASD powder. Because solvent evaporation time is extremely fast (on
the order of seconds), spray drying is particularly advantageous for
preparing ASDs of compounds with poor thermal stability.
There is no limitation to the types of polymers that may be employed
for preparation of ASDs by spray drying. In particular, spray drying
enables the preparation of ASDs in HPMC based polymers, which is
often difcult, if not impossible, to achieve using rotary evaporation or
HME processes. Spray drying also ofers the opportunity to optimize
particle size and bulk powder properties through process parameter
optimization and also through the type of spray nozzle (e.g. two-fuid,
ultrasonic, rotary, and pressure nozzles) [4]. In general, particle size
increases with equipment scale, as a result of larger droplet sizes and
longer drying residence times. This may present difculties during
development, as particle size of the spray dried powder is inherently
changing as the formulation is scaled. This can be especially challenging
during discovery and early development, because smaller batch sizes
lead to inherently small particles (~10 m) which can lead to issues
with fow and compressibility during downstream processing. Issues
with particle size can be largely mitigated via dry granulation of the
spray dried powder, however this adds another relatively complicated
unit operation to the overall process. Another challenge to employing
spray drying during discovery and early stages of development is that
the currently available lab scale spray dryers sufer from poor yield and
generally cannot work on mg quantities of material
Hot Melt Extrusion
Hot melt extrusion (HME) is the most widely used method of
preparation for ASDs for commercial products, because it is
particularly well suited for large scale manufacturing [5,6]. The
preparation of ASDs by HME typically involves the use of twin screw
extruders to mix multiple materials (API, polymer, surfactant) into a
melt which is extruded through a die. The extrudate is then cooled
and either shaped by calendaring or pelletized and milled to a desired
particle size. The fnal milled extrudate is then typically blended
with additional excipients and compressed. Direct shaping to a fnal
dosage form is also possible with calendaring or injection molding
technology. HME is advantageous for commercial manufacturing
because it is a continuous and easily scalable process. Unlike rotary
evaporation and spray-drying, HME does not require the use of organic
solvents, thus it is a green process that reduces cost and alleviates
safety/environmental concerns. Processing must be performed at
temperatures above the Tg of the polymer and high enough for the
API to either melt and/or dissolve into the polymer matrix. HME can
be limited in the ability to process heat sensitive and/or high melting
point drugs and it is generally not amenable for manufacturing small
(mg to g) quantities needed in preclinical development.
selection of Excipients
Polymers
Polymers are critical components in ASDs because they act as carriers
for the drug and they inhibit crystallization in both the dosage form
and in-vivo. By remaining in an amorphous state during dissolution, the
drug can achieve supersaturation and potentially greater absorption,
when solubility is the limiting factor.
In addition to in vivo performance considerations, polymer properties
such as the glass transition temperature (Tg), solubility in organic
solvents, and hygroscopicity must be considered in order to
make the ASD stable and manufacturable. The properties of some
commonly used polymers for preparation of ASDs are summarized
in Table 1. The polymer Tg is an important property to consider
when preparing and selecting an ASD formulation. Polymers with
higher Tg have less mobility, lending to better inhibition of drug

January/February 2011 |

| 69

FormulAtIon DEvEloPmEnt
crystallization. Additionally, the polymer
Tg is particularly important for hot melt
extrusion, as the process must be carried out
above Tg to sufciently mobilize the polymer.
Organic solvent solubility of the polymer is a
critical factor when manufacturing by rotary
evaporation or spray drying to ensure that the
polymer can be fully dissolved at the required
concentration. The hygroscopicity of the
polymer must also be considered, because an
increase in moisture content can negatively
afect physical and chemical stability, and
proper packaging may be needed for ASDs
composed of hygroscopic polymers.
Surfactants
Surfactants are often used as solubilizers or
emulsifying agents in ASDs. Their primary
purpose is to increase the apparent aqueous
solubility and bioavailability of the drug. The
properties of some common surfactants used
in ASDs are listed in Table 2. As with polymers,
solubility in organic solvents is an important
consideration when preparing ASDs from
solvent. In the case of hot melt extrusion,
surfactants can have a plasticizing efect, which
allows processing at lower temperatures.
Organic Solvents
Solvents are necessary when preparing ASDs
by rotary evaporation or spray drying. The
properties of some common solvents used for ASD
preparation are listed in Table 3. Solubility of the
drug typically drives the solvent selection process,
but all components should be completely dissolved
to produce a homogeneous feed solution and a
consistent fnal ASD powder. The solubility of the
components in the chosen solvent must be high
enough to manufacture at a reasonable throughput
(typically > 5% weight of the total solids load).
Water is often employed as a cosolvent for drugs
(e.g. hydrates) that exhibit maximum solubility in
a water-organic solvent mixture. The boiling point
of the solvent is used as a guideline to set process
temperatures in both rotary evaporation and
spray drying processes. Sometimes a system with
multiple organic solvents can be used to improve
the solubility of various components. For GLP/GMP
manufacturing, the ICH limit of the chosen solvent
must be considered and secondary drying is often
necessary to remove residual solvent.
Table 1: Properties of Polymers Commonly Used in ASDs [7]
Polymer Tg (C) Solvent Solubility Hygroscopicity Amenable Methods
of Manufacture
Copovidone 106
Dichloromethane
Ethanol
Methanol
Water
Acetone
<10% @ 50% RH
Rotary Evaporation
Spray Drying
Hot Melt Extrusion
Polyvinyl
caprolactam-
polyvinyl acetate-
polyethyleneglycol
copolymer
70
Water
Ethanol
Methanol Acetone
~5% @ 50% RH
Rotary Evaporation
Spray Drying
Hot Melt Extrusion
PVP
130 (K17)
168 (K30)
Chloroform
Ethanol
Methanol
Water
Acetone
~15% @ 50%RH
Rotary Evaporation
Spray Drying
Hot Melt Extrusion
HPMC 170
Cold Water
Dichloromethane: Ethanol
Dichloromethane: Methanol
Water: Alcohol
<10% @ 50% RH Spray Drying
HPMC P 133 - 137
Acetone: Methanol
Acetone: Ethanol
Methanol: Dichloromethane
2 5% @ 50%RH Spray Drying
HPMC AS 110 - 130
Acetone*
Ethanol:Dichloromethane*
*clear or turbid viscous solution
~3% @ 50%RH Spray Drying
Methacrylate/
methacrylic acid
copolymer
110 - 150
Ethanol, Methanol, Acetone,
Acetone with 3% water
<5% @ 50% RH
Rotary Evaporation, Spray
Drying, Hot Melt Extrusion
Salt/polymorph/cocrystalscreening
Preformulationstudies
Amorphoussoliddispersiondevelopment
Excipientcompatibilityscreens
APIformselectionandoptimization
SolidstateNMRservice
Chiralresolutionbycrystallization
Solidstatemethodsdevelopment,validationandtransfer
BasedinChina,partneringwithpharmaceuticalcompaniestoprovidehighquality
integratedsolidstatechemistrysolutionsforbothAPIandformulationdevelopment
High quality, fast turnaround, cost effective
and fexible to suit all your needs
www.crystalpharmatech.com
Our Services

70 |

|

January/February 2011
FormulAtIon DEvEloPmEnt

Small-Scale ASD Screening


When conducting a screen for ASDs, the primary objective is to
identify a formulation that enables in-vivo exposure of a poorly water-
soluble compound and one that is also stable, both chemically and
physically. For this, a wide range of polymers and polymer-surfactant
systems can be screened. In addition, the drug and surfactant loading
in the ASD can be evaluated for its efects on release behavior and in-
vivo performance, as well as physical and chemical stability. If multiple
combinations of polymers, surfactants, and drug/surfactant loads are
screened, the number of samples can easily range into the hundreds.
For early stage screening, a centrifugal solvent evaporator can be
used to quickly prepare a large number of samples, in parallel, using
mg quantities of material. Samples can be prepared using 96-well
plates for small (mg) scale screening or gram-scale quantities can be
prepared using scintillation vials or small beakers [9]. Small samples
allow for larger, more comprehensive screens to be carried out quickly
while still providing enough material for meaningful characterization.
Potential lead formulations can then be manufactured on a larger
scale for further evaluation, including physical and chemical stability
studies, in-vitro release characterization, and in-vivo studies in animals.
Characterization
Characterization of an ASD is a critical requirement following
preparation in order to be confdent in the performance of the
formulation. Characterization should include analyses of both solid
form and in-vitro API release in aqueous media. Numerous methods
are available for characterization, the more common of which are
described in the following sections and in Table 4.
Solid Form Evaluation
Of primary importance is the identifcation of residual crystalline
character as this signals potential long-term physical instability of
the amorphous API. Polarized light microscopy (PLM) and powder
X-ray powder difraction (PXRD) are rapid, non-destructive techniques
employed as an early screen for crystallinity. Individual crystals may
be identifed via microscopy while PXRD difractograms provide
information regarding the gross amorphous or crystalline character
of the solid. Data from both methods provide greater insight when
combined with more sensitive thermal methods such as diferential
scanning calorimetry (DSC).
DSC is employed to study transitions in the solid state as a function
of temperature. Most often, ASDs are evaluated for the presence of
melt endotherms and Tg. Observation of melt endotherms confrms
the presence of crystalline components. Tg is an important descriptor
that can provide insight into the long term stability of the ASD. Since
a higher Tg generally indicates better stability, a single, high Tg value
is desired for a particular ASD. Tg generally decreases with increased
moisture uptake, which makes the API more prone to crystallization
at higher RH conditions. Multiple Tg values indicates heterogeneity
in the system and an increased potential for phase separation within
the solid leading to crystallization. Thermogravimetric Analysis (TGA)
is a thermal method that is complementary to DSC. TGA is employed
to study weight loss from an ASD as a function of temperature and
this technique is typically employed to roughly quantitate amounts
of residual water or organic solvent present in the ASD. It is possible
to determine the identity of materials lost during heating by coupling
the TGA to a mass spectrometer (TGA-MS). These data may then be
utilized to adjust processing or storage parameters to minimize the
amount of plasticizing solvents which may lead to physical instability
for the ASD. Dynamic vapor sorption can also be performed to
understand the hygroscopic tendencies of the ASD powder.
Many other analytical methodologies are available to further
characterize ASDs, although these are typically less common. Some
of the other techniques include solid-state NMR spectroscopy [10],
Raman spectroscopy [11], infrared spectroscopy [12], and isothermal
microcalorimetry [13].
Table 4: Common Techniques Employed for ASD Solid Form
Characterization
Technique Output Observations Analysis
Microscopy Photmicrograph Birefringence
Crystalline or amorphous
character based on presence
or absence of birefringenge
PXRD
X-Ray
Difractogram
Presence or Absence
of Observable, Intense
Peaks
Crystalline or amorphous
based on characteristic
presence or absence of
well defned refections in
difractogram
TGA Thermogram Weight Loss
Residual water or organic
solvent
DSC Thermogram
Tg, single
Homogeneous amorphous
phase
Tg, multiple Phase Separation
Endothermic Events Desolvation or Melting
Exothermic Events Potential recrystallization
Table 2. Properties of Surfactants Commonly Used in ASDs [7]
Surfactant HLB T
m
(C) Solvent Solubility
Vitamin E Polyethylene Glycol Succinate 13.2 37 - 41
Water
Acetone
Sorbitan Monostearate 60/80 4.3 53 - 57 Most organic solvents
Polysorbate 20 16.7 n/a
Ethanol
Water
Polysorbate 80 15.0 n/a
Ethanol
Water
Polyoxyl 40 Hydrogenated Castor Oil 14 - 16 30
Chloroform
Ethanol
Acetone
Water
Table 3. Properties of Organic Solvents Commonly Used for
Preparation of ASDs [8]
Solvent Boiling Point (C) Flash Point (C) ICH Class
Ethanol 78 13 II
Methanol 65 12 III
Acetone 57 17 III
IPA 83 12 III
Tetrahydrafuran 66 -14 III
Dichloromethane 40 None II
Published 11/10 PATH0153R0
US Headquarters
Patheon Inc.
PO Box 110145
Research Triangle Park, NC 27709-5145
USA
P: +1 919 226 3200
F: +1 919 474 2269
www.patheon.com
European Headquarters
Patheon International AG
Lindenstrasse 14
6340 Baar
Switzerland
P: +41 41 766 2580
F: +41 41 766 2581
www.patheon.com
Why limit your compound to just one
bioavailability enhancement technology?
One price, multiple technologies fast results.
Call +1 866-PATHEON (+1 866-728-4366) or email doingbusiness@patheon.com
the first fixed-price, multi-platform scalable solution to increased bioavailability
parallel drug formulation screening including expert scientific analysis
SoluPath the fast path to phase I trials and commercialization

72 |

|

January/February 2011
FormulAtIon DEvEloPmEnt

Long term stability of ASDs should be evaluated due to the increased


risk of both physical and chemical instability associated with
amorphous solids. Accelerated stability studies should be conducted
under stressed conditions (e.g. open dish at 40C/75% RH, 60C/75%
RH, etc.) to understand both the physical stability of the ASD and any
increased risk of chemical reactivity in the presence of excipients.
Photostability should also be examined to determine whether special
packaging is required to prevent photochemical reactions in the
amorphous state. Understanding the stability of an ASD allows one to
reformulate as necessary by varying drug load, by adding antioxidants,
or by selecting alternative polymers/surfactants to maximize stability.
In-vitro Release
Release of API from an ASD may be studied by a variety of in-vitro
methods. One way to evaluate release from ASDs is by simple
powder dissolution. This is performed by transferring a known mass
of material into a known volume of biologically relevant dissolution
medium (e.g. simulated gastric/intestinal fluid) under constant
stirring. Solution concentrations are measured as a function of time
to generate a concentration versus time release profile for the API.
Solution concentrations in significant excess of the equilibrium API
solubility in the particular medium should be targeted to test the
ability of the ASD to achieve and maintain supersaturation.
However, it is desirable to be as biorelevant as possible to predict
in-vivo performance. More dynamic techniques, such as the artifcial
stomach duodenum, and other multicompartmental dissolution/
release methods, should be employed to understand API release from
ASDs [3,14]. These methods are used to understand complex release
phenomena and to begin building in-vitro/in-vivo correlations. A
dynamic dilution scheme reported by Gao et al, was recently shown
to provide valuable inputs for predictions of pharmacokinetics (PK).
Dosing Considerations
Since ASDs are inherently metastable systems, it is important to
consider the implications of the dosage form and dosing conditions
on physical/chemical stability, manufacturability, and in-vivo
performance. Aqueous suspensions enable dosing of higher ASD
amounts and they are therefore ideal for dosing to animals for
toxicological evaluation. Care must be taken to ensure that the ASD
does not crystallize in the aqueous suspension during preparation and
dosing, which could compromise in-vivo performance. Gelling and/or
foaming can occur when suspending an ASD in aqueous solution. This
issue may be overcome through optimization of the ASD loading in
the suspension and/or addition of anti-gelling/foaming agents. When
dosing smaller animals by oral gavage, the particle size of the ASD
in aqueous suspension must be small enough to pass through the
gavage tube.
Hard gelatin capsules (HGC) are ideal for dosing of the dry ASD to
larger animals and humans. If long term stability is required, the
physical and chemical compatibility of the ASD with the capsule shell
must be considered, especially for hygroscopic polymers which may
dry out or cause the HGC to swell depending on the RH conditions.
Tablets are generally the preferred dosage form for ASD formulations
of commercial products. Formulation of the ASD intermediate with
secondary excipients (e.g. fllers, disintegrants, lubricants, glidants) is
typically required in order to optimize the fow and compressibility
of the ASD formulation to enable manufacturability of the fnished
tablet. Thus, the ASD intermediate must be physically and chemically
compatible with the API/ ASD powder. The particle size of the ASD
powder is also an important consideration for tablet formulations,
as the particle size can afect the manufacturability (i.e. fow and
compressibility) as well as the dissolution/release rate of the API from
the tablet matrix. ASDs made by HME or rotary evaporation processes
are typically milled to the optimal particle size for tableting. The
desired particle size of spray dried ASDs may be obtained through
process optimization, especially on large scale spray dryers. Spray
dried powders made on a smaller scale during early development
tend to have inherently small particle size, thus these powders may
require dry granulation (roller compaction, milling) in order to obtain
the desired particle size for optimal tablet properties.
Conclusions
We have outlined the fundamentals of preparing, screening,
characterizing, and dosing amorphous solid dispersions. Using
these methods we have successfully delivered poorly soluble drug
candidates for both preclinical and clinical studies and we believe that
the use of ASD technologies will continue to increase. As the science
of this drug delivery approach evolves and as new excipients become
available, formulators will be able to design ASD systems that are even
more sophisticated and that will ultimately get new medicines to
patients faster.
references
1. C. J. Lipinski, Pharmacol. Toxicol. Methods, Drug-like properties and
the causes of poor solubility and poor permeability,2000, 44, 235-249.
2. E. H. Kerns, J. Pharm. Sci.,High-throughput physicochemical profiling
for drug discovery, 2001, 90, 1838-1858.
3. Gao, Y., Carr, R.A., Spence, J.K., Wang, W.W., Turner, T.M.; Lipari,
J.M.; Miller, J.M., pH-Dilution Method for Estimation of Biorelevant
Drug Solubility along the Gastrointestinal Tract: Application to
Physiologically Based Pharmacokinetic Modeling, Molecular
Pharmaceutics, in press.
4. Dobry, D.E., Settell, D.M., Baumann, J.M., Ray, R.J., Graham, L.J.,
and Beyerinck, R.A., A Model-Based Methodology for Spray-Drying
Process Development, Pharm Innov. 2009, 4, 133142.
5. Breitenbach, J.; Maegerlein, M., Melt-extruded molecular dispersions,
Drugs and the Pharmaceutical Sciences, 2003, 133, 245-260.
6. Breitenbach, J., Melt extrusion can bring new benefits to HIV
therapy: the example of Kaletra tablets, American Journal of Drug
Delivery, 2006, 4, 61-64.
7. Handbook of Pharmaceutical Excipients, 6th Edition, Edited by Rowe
R.C., Sheskey, P.J., and Quinn, M.E., Pharmaceutical Press, 2009.
8. Miller J.M., Blackburn A.C., Macikenas D., Collman B.M., and
Rodrguez-Hornedo N., Solvent Systems for Crystallization and
Polymorph Selection, in Solvent Systems and Their Selection
in Pharmaceutics and Biopharmaceutics, AAPS Biotechnology:
Pharmaceutical Aspects, Volume 6, 2007.

January/February 2011 |

| 73

FormulAtIon DEvEloPmEnt
9. Shanbhag, A., Rabel, S., Nauka, E., Casadevall, G., Shivanand, P.,
Eichenbaum, G., Mansky, P., Method for screening of solid
dispersion formulations of low-solubility compounds-Miniaturization
and automation of solvent casting and dissolution testing
International Journal of Pharmaceutics, 2008, 351, 209-218.
10. Lubach, J.W., Munson, E.J., Solid-State NMR Spectroscopy, in
Polymorphism: in the Pharmaceutical Industry, Edited by R. Hilfkiker,
2006, Wiley-VCH, 81-93.
11. Breitenbach, J., Schrof, W., Neumann, J., Confocal Raman-
Spectroscopy: analytical approach to solid dipsersions and mapping
of drugs, Pharm. Res., 1999, 16, 1109-1113.
12. Broman, E., Khoo, C., Taylor, L.S., A comparison of alternative polymer
excipients and processing methods for making solid dispersions of a
poorly water soluble drug, Int. J. Pharm., 2001, 222, 139-151.
13. Sebhatu, T., Angberg, M., Ahlneck, C., Assessment of the degree of
disorder in crystalline solids by isothermal microcalorimetry, Int. J.
Pharm., 1994, 104, 135-144.
14. Alonzo, D.E., Zhang, G.G.Z., Zhou, D., Gao, Y., Taylor, L.S.,
Understanding the Behavior of Amorphous Pharmaceutical Systems
during Dissolution, Pharmaceutical Research, 2010, 27, 608-618.
Author Biographies
Brian Padden is Section Manager of Pharmaceutics
at Abbott Laboratories. He holds B.A. degrees in
physics and chemistry from Saint Marys University
(Winona, MN), and M.S. and Ph.D. degrees in chemistry
from the University of Minnesota. Dr. Padden started
his career at the Schering-Plough Research Institute, where he was
responsible for solid-state method development, GMP validation, and
technology transfer to international manufacturing sites. Some of the
commercial products that he contributed to during that time include
Claritin, Clarinex, Asmanex, Nasonex, Noxafl, and Zetia. Dr.
Padden then served in positions of increasing responsibility in the areas
of preformulation and discovery support. In 2006, he joined Abbott
and in his current role he is responsible for advancing the pipeline
through material-sparing characterization and enabling preclinical
formulations, in the therapeutic areas of oncology, neuroscience, pain,
and dyslipidemia. He has received many technical awards, including the
Abbott Presidents Award in 2009, and he is a certifed Lean Six Sigma
Black Belt. Dr. Padden has published and presented dozens of papers,
posters, and invited talks, and he is also a board member of the NSF
Center for Pharmaceutical Development.
Jonathan Miller is a Principal Scientist in the Global Formulation
Sciences - Solids group at Abbott Laboratories. He is also Adjunct Assistant
Professor in the Department of Pharmaceutical Sciences at the University
of Michigan. He has over 12 years of industrial experience both at Pfzer and
now Abbott Labs, building broad expertise in the areas of pre-formulation,
solid form screening/selection, formulation, biopharmaceutics, and
material science. He has authored over 20 scientifc publications and
is an inventor on more than 10 patents and patent applications. In his
current role at Abbott, he is responsible for the development of enabling
formulations for poorly soluble compounds including amorphous solid
dispersion formulations, lipid based drug delivery systems, and nano-
particles. Dr. Miller holds a Ph.D. in Pharmaceutical Sciences and an
M.S. in Pharmaceutical Engineering from the University of Michigan. He
obtained his B.S. in Biochemistry from Bowling Green State University.
Timothy Robbins is the Operations Manager of the Chemical Pilot
Plant at Abbott Laboratories. Dr. Robbins joined Abbott in 1993 after
completing post-doctoral work at the University of California, Los Angeles.
He has over 17 years of chemical research and scale-up experience. He
has 12 scientifc publications and is an inventor on more than 7 patents
and patent applications. In his current role at Abbott, he is responsible
for the running of the Chemical Pilot Plant and Kilo Lab. Prior to joining
the Chemical Pilot Plant, Tim worked on a number of projects as a process
research chemist within the API Process R&D organization. Dr. Robbins
holds a Ph.D. in Organic Chemistry from the Unviversity of Nevada-Reno.
He obtained his B.S. in Chemistry from Olivet Nazarene University.
Philip Zocharski is a Senior Scientist in the Pharmaceutics group at Abbott
Laboratories. Over his 12 year career in the pharmaceutical industry,
Philip has focused his research and development eforts in the areas of
pre-formulation and biopharmaceutics as a colleague of Parke-Davis/
Pfzer and Abbott Labs. In his current role, he applies biopharmaceutics
principles and a wide array of drug delivery technologies including
amorphous solid dispersions, nanoparticles, and lipids to address
the specifc needs of teams in early Discovery. Philip holds an M.S. in
Pharmaceutical Sciences from the University of Michigan. He obtained his
B.S. in Chemistry from Michigan State University.
Leena Prasad is a Scientist I in the Global Formulation Sciences Solids
group at Abbott Laboratories. She graduated in 2005 from the University
of Illinois at Champaign/Urbana with a B.S. in Mechanical Engineering.
She joined Abbott in July of 2007. Since joining Formulation Sciences,
she has actively been involved with various projects requiring solubility
enhancement technologies and has worked with the groups solid
dispersion research team.
Julie Spence is a Scientist I in the Pharmaceutics group at Abbott
Laboratories. She earned her B.S. in Chemical Engineering in 2001 from
the University of Michigan and her M.E. in Pharmaceutical Engineering
in 2002, also from the University of Michigan. Prior to joining Abbott in
2007, Julie worked as a Scientist at the Pfzer Ann Arbor, MI labs. (2002
- 2007). Over the course of her career, Julie has gained expertise in the
areas of physicochemical characterization, dermatological drug delivery,
biopharmaceutics, and solid form screening/selection. Julies current
responsibilities include developing enabling formulations for toxicology
studies.
Justin Lafountaine is an Associate Scientist II in the Global Formulation
Sciences Solids group at Abbott Laboratories. He graduated in 2007
from the University of New South Wales with a B.S. in Nanotechnology.
After graduation, Justin worked briefy at Pharmaform before joining the
Global Formulation Sciences group in September of 2007. Since then, he
has actively been involved with expanding the groups solid dispersion
capabilities and implementing Process Analytical Technology.

74 |

|

January/February 2011
PFs / E&l

Introduction
Preflled glass syringes (PFS) are increasingly becoming a container of
choice for storing and administering therapeutic protein products to
patients [1]. The PFS is a convenient and reliable system for injection
compared with the more traditional method of transferring, measuring,
and delivering a dose from a vial containing liquid or reconstituted
lyophilized powder. In addition, the PFS can deliver a controlled
volume minimizing drug waste associated with vial overflls.
PFS is a primary container and its compatibility with the drug
needs to be addressed for ensuring patient safety and drug quality
[2, 3]. Safety of PFS is the responsibility of drug manufacturers
and understanding PFS extractables/leachables is important for
assessing syringe compatibility. Identifcation and quantitation of
extracted/leached chemicals is critical for assessing toxicological
and drug quality risks. A high extractable/leachable risk assessment
may simply disqualify a syringe and eliminate the need to conduct
further resource intensive qualifcations assessments such as stability,
particle formation, container closure integrity, break loose extrusion,
and others. Understanding extractable/leachable contributes to the
PFS knowledge space and may also serve to create collaboration
opportunities with suppliers for reducing/eliminating leachables
and improving PFS systems [4]. This paper describes extractables/
leachables from syringes and its implication on biological products.
Extractables and leachables
Extractables are chemicals that migrate from the product-contact
component into a solvent at accelerated conditions (such as heat, time,
pH, ionic strength, organic solvent content). Leachable are chemicals
that migrate from the product-contact component into a formulated
drug during normal storage/usage conditions.
Information regarding leached chemicals from components is not
known or readily available from the supplier. Goals of extractable
studies are to generate, identify, and predict leachables. Extraction
under appropriate solvent, temperature, and exposure conditions can
generate representative leachables and in large enough quantities
to facilitate structure elucidation analysis [5, 6]. Assembled syringes
and individual components can be extracted with various solvents
(water and water/organic mixtures, pH, ionic strength) at elevated
Yasser Nashed-Samuel, Dengfeng Liu,
Kiyoshi Fujimori, Lourdes Perez & Hans Lee, Ph.D.
Department of Formulation and Analytical Resources
Amgen Inc.
Extractable and Leachable
Implications on Biological
Products in Preflled
Syringes
Daikyo Crystal Zenith

and Flurotec

are registered trademarks of Daikyo Seiko, Ltd. Crystal Zenith

and Flurotec

technologies are licensed from Daikyo Seiko, Ltd.


West and the diamond logo are trademarks of West Pharmaceutical Services, Inc. in the United States and other jurisdictions.
Copyright 2011 West Pharmaceutical Services, Inc.
Breakage. Functional performance. Aggregation.
These issues and more can affectyour product
and thus your top and bottom line. Glass
syringes may be at the heartof theproblems.
Made of a proprietary cyclic olefn polymer,
theDaikyo Crystal Zenith Insert Needle
Prefllable Syringe System provides an
innovative solution to the limitations of
glass syringes, especially with sensitive
biologic drug formulations.
System advantages include:
Highbreak-resistance
Highestqualityassuredby
100%visioninspection
Superiorfunctionalperformanceand
barrierprotection with Flurotec

flm
Nosiliconeoilonthesyringe
barrel or piston
Notungstenoradhesive
Consistentfunctionalitywhenused
inauto-injectorsystems
Advance Your Delivery System
with Daikyo Crystal Zenith


Insert Needle Prefllable
Syringe Technology
Minimize Risk. Maximize Value. Enhance Delivery.
#6001
Contact West today.
NorthAmerica+1800-345-9800
Europe +4924037960
AsiaPacifc +6568605879
www.WestPFSsolutions.com
Get the free mobile app at:
http://gettag.mobi
6001 1ml CZ Syringe PFS ad.indd 1 2/7/2011 11:43:17 AM

76 |

|

January/February 2011
PFs / E&l

temperatures for a specifed duration [5-8]. The identifed extractables


provide insight on predicting leachables. Leachables can be identical
or a subset (oxidation, derivative, etc.) of extractables, or can form
adduct products with other leachables, excipients, and proteins.
Extractables information is benefcial since analyzing leachables
directly in the formulated drug is challenging due to low leachable
concentrations (ppm/ppb range) and matrix inference from protein
and excipients. Also categorizing foreign chemicals in drug products
as syringe component or drug process impurity related is inconclusive
when analyzing leachables directly. Extractables data can determine
the source of the leachables.
Analytical techniques
Analytical characterization and quantitation techniques associated
with organic and inorganic chemicals are commonly used to
analyze extractable/leachables [9]. Gas chromatography mass
spectrometry (GCMS), solid phase micro extraction (SPME)-GCMS,
liquid chromatography mass spectrometry (LCMS), high performance
liquid chromatography (HPLC), and nuclear magnetic resonance
(NMR) spectroscopy techniques are used to analyze unknown
organic extractable compounds. Inductively coupled plasma mass
spectrometry (ICPMS) can be used to identify and quantitate inorganic
metallic elements. Evaporative light scattering detection (ELSD) can
be supplemental to HPLC and suitable for oligomers/polymers and
non-chromophores molecule analysis.
PFs Components, leachables,
and Implications
Sources of leachables can be from the PFS components (glass barrel,
stainless steel hypodermic needle, rubber needle shield, silicone
oil lubricants, and fuoropolymer coated rubber plunger) and less
obvious sources are from [8, 10] residues from processing tools [11-13]
and additives for attaching the needle to the barrel [4]. The impact
of leachables on therapeutic proteins can be related to aggregation,
particle formation, and/or product quality issues such as reaction with
the formulation or protein. Many of these negative attributes may not
be observed in non PFS containers possibly due to short contact times
during the uptake and delivery from vials using non PFS. However,
the shelf-life for biological therapeutics in PFS can be greater than one
year allowing leachables to accumulate and potentially interact with
drug formulations [14].
The impact from leachables may not be universally observed across
all formulated products in a pipeline since formulation factors (pH,
ionic strength, surfactants, excipients, etc.) and/or protein attributes
may vary.
glass
Type I borosilicate glass is commonly used to make preflled syringes
and contains various inorganic oxides such as boron, silicon, calcium,
sodium, potassium, iron, and aluminum [15]. These inorganic
oxides may not pose a direct toxicological risk but migration of glass
components from ion-exchange, glass dissolution, pitting, stress,
surface layer exfoliation, weathering, and/or erosion corrosion efects
may lead to particle formation. [7,15].
rubber materials
(Plunger and needle shield)
The plunger and needle shield are composed of rubber related
materials. The product contact surface of the plunger is laminated with
a fuoropolymer type flm and the needle shield is unlaminated. Contact
between the formulated drug with the plunger and shield may result
in leached rubber additives or bromine related compounds. Leached
rubber agents have been proposed to cause adverse patient efects [10].
hypodermic needle
The attached hypodermic needle is made of stainless steel. Inorganic
metals used in the stainless steel formulation such as Fe, Cr, Mn, Ni,
and Mo may leach from the needle. Based on our experience, metals
leach at low levels and are not a direct toxicological concern. However
under specifc conditions and concentrations, metals such as Mn may
catalyze oxidization reactions on proteins [16].
Adhesive
UV activated adhesives composed of organic and oligmeric/polymeric
materials are commonly used to bind the stainless steel hypodermic
needle to the glass barrel [4]. Controlling the adhesive formulation,
application, activation/curing, residues, and clean up processes are
important steps for preventing/reducing these materials from coming
in contact and leaching into the drug formulation.
While evaluating PFS models from various vendors, several organic,
oligomeric, and acrylate related materials consistent to those used
in the adhesive industry were extracted and characterized [17].
These adhesive residues did leach into various formulated protein
products and did not appear to induce precipitation, aggregation, or
particles. The impact on drug quality was a concern since acrylates are
reactive toward protein under certain conditions [18]. Evaluation of a
formulated protein stored in syringes containing endogenous levels
of adhesives at 37C for 45 days led to adduct formation. Irreversible
addition of acrylates with the formulated protein was observed at
multiple lysine, histidine, and N-terminus sites with 0.02% of amino
acid sites being modifed [19].
SOME SKILLS ARE ESSENTIAL
SOME PRODUCTS AS WELL
AGUETTANT has designed a
unique polypropylene prelled
syringe to improve safety and
quality of care whilst optimizing
cost levels.
Ready to use
Prelled, preassembled and presented
in a blister pack
Sterility guaranteed inside & outside
through terminal sterilization
Integrity
Guaranteed by the frangible obturator
Tamper evident
Patented opening system with
pre-perforated label
Needle free
Secure connection with its universal
Luer Lock

INNOVATION FOR ALL
FOR YOUR PREFILLED DEVELOPMENT
Come and visit us at BIOMEDevice on booth 345, Boston Convention & Exhibition Center, April 6-7th, 2011
AGUETTANT - 1 Rue Alexandre Fleming - 69007 LYON - France www.aguettant.com
Tl: +33 (0)4 78 61 51 41 - Fax: +33 (0)4 78 61 09 35 - Email : business.development@aguettant.com

78 |

|

January/February 2011
PFs / E&l

silicone oil
Silicone oil is applied to coat the barrel, plunger, and needle exterior.
The most common form of silicone oil used in medical applications
is polydimethylsiloxane and functions as a lubricant allowing the
plunger to glide smoothly within the barrel to expel the drug. As
the demand for preflled syringe and automated injection devices
increases, so does the importance of understanding silicone oil.
Functionally, silicone oil is not a major concern for manual injections
since a nurse or doctor is capable of applying the necessary force to
push the plunger to the end point. However, a spring can only provide
a fxed amount of force and any unanticipated friction may cause the
plunger to stall before complete drug delivery.
Although silicone oil is considered inert and insoluble in water, it
may interact with the formulation causing protein aggregation
[20,21], droplets, and particles [22]. Ideally the silicone oil application
process should balance the need to minimize the undesired drug
quality attributes and to provide sufcient lubrication. The silicone
oil distribution is non-uniformly distributed with the least amount
located near the needle end of the syringe (Figure 1) [23].
syringe tool tungsten Pin
Less obvious sources of PFS extractables are contamination from tools
that were used to manufacture and process syringes. Glass syringes
are made from cutting and molding glass tubing at high temperatures.
Heated glass is in contact with various processing tools during the
process. The barrels inner channel cavity for holding a stainless steel
needle is formed at approximately 1,200 C using a tungsten pin.
Tungsten is commonly used due its heat resistance and relative high
melting temperature. However at temperatures greater than 150 C,
tungsten oxidizes in the presence of air and leaves a white tungsten
containing residue within the syringe (Figure 2). These residues may
survive the syringe washing step and may contact the drug upon
syringe flling and storage. The tungsten oxide residues did not cause
protein precipitation above pH 5, but caused protein aggregation
below pH 5 [12, 24]. Below pH 5, ppm low amounts (parts per million,
ppm) of tungsten oxide formed large tungstate polyanions which did
aggregate protein at low ppm levels.
syringe tool Polymeric Pin
Polymeric nylon pins are used to transport glass syringe barrels on an
assembly line. These reusable pins (approximately 0.5 x 6 cm) ft within
the hot syringe (Figure 3). Abnormal heat exposure or extensive pin
usage may lead to pin wear and tear. Pin residues exposed to heated
glass syringes may adhere to the inner syringe wall and survive wash
and rinse procedures. During a visual inspection of a flled syringe
product a black residue was observed (Figure 4) and later identifed
by LCMS and FTIR analysis as containing nylon related species [11].
Substances from the black residue did leach into the drug formulation
Figure 1. Silicone oil gradient in an empty PFS [23].
Figure 2. Tungsten oxides inside the syringe near the hypodermic
needle-barrel zone [24].
Figure 3. Polymeric pin and a polymeric pin inserted into a syringe.

January/February 2011 |

| 79

PFs / E&l
and analysis confrmed the leachables and solid black residue matched
those of the nylon pin used by the syringe manufacture. The syringe
manufacturer was notifed and has implemented measures to prevent
future occurrences.
Conclusion
PFS components and residues from processing tools may leach organic
and inorganic chemicals into formulated drugs. Leachable information
is often not readily available from the syringe manufacturer prompting
drug manufacturers to initiate extractable/leachable investigations.
Leachable from PFS may or have contributed to safety concerns
related to protein aggregation, particle formation, and toxicological
risk factors. Identifying extractables and leachables provide key
information enabling safety assessments that address toxicology and
drug quality impact for evaluating PFS.
Acknowledgements
Authors would like to thank Joseph Phillips and David Brems for their
eforts and useful discussions.
references
1. Thompson, I. New-Generation Auto-Injectors: Completing the
Scale of Convenience for Self-injection. Drug Delivery Report. 2005,
Autumn/Winter, 47-49.
2. Markovic I. Risk Management Strategies for Safety Qualification of
Extractable and Leachable Substances in Therapeutic Biologic Protein
Products. Am Pharm. Rev. 2009, 12(4) 96-101.
3. Guidance for Industry. 1999. Container closure systems for packaging
human drugs and biologics. Rockville, MD: US Department of Health
and Human Services, Food and Drug Administration.
4. Sardella, A. Fine tuning of process parameters for improving
biocompatibility of prefillable syringes. Ondrugdelivery. 2010,
(January) 18-22.
5. Jenke, D. R. Evaluation of model solvent systems for assessing the
accumulation of container extractables in drug formulations. J.
Pharm. Sci. 2001, 224 (1-2), 5160.
6. Jenke, D. R. Linking extractables and leachables in container/closure
applications. PDA J. Pharm. Sci. Technol. 2005, 59 (4), 265281.
7. Borchert, S.J.; Ryan, M.M.; Davidson, R.L.; Speed, W. Accelerated
extractable studies of borosilicate glass containers. J. Parenter. Sci.
and Technol. 1989, 43(2) 67-79.
8. Wakankar, A.A.; Wang, J. Y.; Canova-Davis, E.; Ma S.; Schmalzing,
D.; Grieco, J.; Milby, T.; Reynolds, T.; Mazzarella, K.; Hoff, E.;
Gomez, S.; Martin-Moe, S. On Developing a Process for Conducting
Extractable-Leachable Assessment of Components Used for Storage
of Biopharmaceuticals. J. Pharm. Sci. 2010, 99(5), 22092218.
9. Wang, Q. Selection of Analytical techniques for pharmaceutical
leachables studies. Am Pharm. Rev. 2005, 8(6) 38-44.
10. Jenke, D. Suitability-for-Use Consideration for Prefilled Syringes.
Pharm. Technol. 2008, April 1 issue.
11. Nashed, Y.; Torraca, G.; Liu, D.; Fujimori, K.; , Zhang, Z.; Wen, Z.; Lee,
H. Identification of an Extraneous Black Particle in a Glass Syringe:
Extractables/Leachables Case Study. PDA J. Pharm. Sci. Tech. 2010,
64, 242-248.
12. Jiang, Y. ; Nashed-Samuel, Y. ; Li, C. ; Liu, W. ; Pollastrini, J. ; Mallard,
D. ; Wen, ZQ. ; Fujimori. K. ; Pallitto, M. ; Donahue, L . ; Chu, G. ;
Torraca, G. ; Vance, A. ; Mire-Sluis, T. ; Freund, E. ; Davis, J. ; Narhi, L.
J. Pharm Sci. 2009, 98(12), 4695-710.
13. Liu, W.; Swift, R.; Torraca, G. ; Nashed-Samuel, Y. ; Wen, ZQ. ; Jiang,
Y. ; Vance, A. ; Mire-Sluis, A. ; Freund, E. ; Davis, J. ; Narhi, L. PDA J.
Pharm. Sci. Tech. 2010, 64(1), 11-19.
14. Hung G.W.; Nunez L.J.; Autian J. Correlation of kinetic parameters
and thermal behavior of segmented polyurethane elastomers with
biological responses. J. Pharm. Sci. 1975, 64, 14921497.
15. Walther, M.; Rupertus, V.; Seemann, C.; Brecht, J.; Hormes, R.; Swift,
R.W. Pharmaceutical Vials with Extremely High Chemical Inertness.
PDA J. Pharm. Sci. Technol. 2002, 56(3), 124-129.
16. Deman, J. M. Principles of food chemistry 3rd edition. (1999) Aspen
Publishers. ISBN: 0-8342-1234-X, 131-132.
17. Nashed-Samuel, Y. Extractable and Leachable Implications on
Biological Products in Prefilled Syringes. PDA/FDA Joint Regulatory
Conference, September 13-16, 2010.
18. Potter, D. W.; Tran, T. Rates of Ethyl Acrylate Binding to Glutathione
and Protein. Toxicology Letters. 1992, 62, 275-285.
19. Liu, D.; Nashed-Samuel, Y.; Bondarenko, P. V.; Brems, D. N.; Ren, D.
Interactions Between Therapeutic Proteins and Acrylic Acid Leachate.
In preparation.
20. Jones, L. S., Kaufmann, A., and Middaugh, C. R. Silicone Oil Induced
Aggregation of Proteins. J. Pharm. Sci. 2005, 94(4), 918-927.
21. Thirumangalathu, R., Krishnan, S., Ricci, M.S., Brems, D.N., Randolph,
T.W., and Carpenter, J. F. Silicone Oil- and Agitation-Induced
Aggregation of a Monoclonal Antibody in Aqueous Solution. J.
Pharm. Sci. 2009, 98(9), 3167-3181.
22. Markovic, I. Challenges Associated with Extractable and/or Leachable
Substances in Therapeutic Biologic Protein Products. Am. Pharm. Rev.
2006, 9(6), 20-27.
23. Lee, H.; Liu, D.; Fujimori, K.; Perez, L.; Nashed-Samuel, Y. Unpublished
results. 2006.
Figure 4. Two black particles observed inside a pre-flled syringe.
Larger particle is approximately 300 microns [11].
80 |

|

January/February 2011
PFs / E&l

24. Lee, H.; Nashed-Samuel, Y.; Fujimori, K.; Liu, D.; Perez, L. Tungsten
Leaching from Prefilled Syringes and Impact on Protein Aggregation.
Poster presented at the PDA Extractables/Leachables Forum:
Confronting Extractables and Leachables Issues in an Evolving
Industry; Bethesda, Maryland, November 68, 2007.
Biography
Yasser Nashed-Samuel, Ph.D., is currently a principal scientist at
Amgen (Thousand Oaks, CA), Process and Product Development, R &
D organization. Since joining Amgen in 2003, he established and led
the leachables and extractables (L/E) efort. The L/E group engages in
assessing product contact for manufacturing equipment, bulk containers,
primary delivery containers and devices and incident investigations for
both clinical and commercial products.
Dengfeng Liu, Ph.D. is a senior scientist in Process & Product Development
at Amgen. He joined Amgen in 2006. Dengfeng leads structure elucidation
for L/E by mass spectrometry and NMR spectroscopy. He conducted
research on the interactions between small molecules therapeutic
proteins.
Kiyoshi Fujimori. Graduated University of California, Los Angeles in 1996
with BS in biochemistry. Initially worked as peptide chemist at Bachem
for six years. Joined Amgen in 2004 and currently studying leachable and
extractable as associate scientist in Product Contact Assessment team of
Process and Product Development.
Lourdes Perez is currently a Sr. Associate at Amgen (Thousand Oaks,
CA), Process and Product Development, R & D organization. Since joining
Amgen in 2006, she has supported leachables and extractables (L/E)
projects, incident investigations, and primary delivery containers and
devices utilized for clinical and commercial products.
Hans Lee, Ph. D. has been with Amgen since 2003 in the Process and
Product Development organization and is responsible for extractables/
leachables activities related to assessing product contact for
manufacturing/infusion/device equipment and bulk/primary containers
for clinical and commercial products. Hans has a Ph.D. in inorganic
chemistry at UCLA.
300+
CHARLOTTESPREMIER
BOUTIQUEHOTEL
MODERNGUESTROOMS
MEETING&EVENTROOMS
C
E
N
T
R
A
L
L
Y
L
O
C
A
T
E
D

I
N
D
O
W
N
T
O
W
N

C
H
A
R
L
O
T
T
E
6200
#OFGUESTSOUR
MEETINGSPACE
CANACCOMMODATE
555 South McDowell Street
Charlotte, NC 28204
RESERVATIONS: 888-664-6835
blakehotelnc.com
T
H
E
FITNESS
CENTER
What is your companys primary business? (Check one only)
1 Pharmaceutical Manufacturing 8 Bulk Materials/Nutritionals
2 Biopharmaceutical Manufacturing 9 Specialty Ingredients/API
3 Contract Pharmaceutical Manufacturing 10 Government
4 Medical Device/Instrument Manufacturing 11 College/University
5 Clinical Pharmaceutical Services 12 Contract Labs
6 Contract Pharmaceutical Research 99 Other (please specify below)
7 Pharmaceutical Engineering/Consultant Services

What is your job function? (Check one only)
A Research & Development E Lab Management J Purchasing
B Quality Control/Assurance F Project Management K Engineering
C Validation G IT/Data Management L Corporate Management
D Production Manufacturing H Regulatory Afairs Z Other (please specify below)
Areas of Interest? (Check all that apply)
1 Analytical Methods 12 Injectables 23 Process Control
2 Aseptic Processing 13 Lipids 24 Single Use Technology
3 Bioprocessing 14 Lyophilization 25 Solid Dosage Formulations
4 Chirality 15 Microbiology 26 Thermal Analysis
5 Controlled Release 16 Molecular Imaging 27 Validation
6 Dissolution 17 NIR 28 Liquid Chromatography
7 Drug Delivery 18 Parenteral Manufacturing 29 Mass Spectroscopy
8 Excipients 19 Particle Sizing 30 Raman
9 Extractables/Leachables 20 PAT/QBD 31 SFC
10 Filtration 21 Peptides 99 Other (please specify below)
11 HPLC 22 Pharma IT
Which tradeshows/conferences do you plan to attend? (Check all that apply)
A PITTCON E INTERPHEX Puerto Rico J Joint Molecular Imaging Conference/WMIC
B EAS F Controlled Release Symposium K PDA
C IFPAC G HPLC L ISPE
D INTERPHEX H FACSS M ASMS
N AAPS
All of the above questions must be answered to qualify for your free subscription. Free subscriptions are available within the
United States and Canada. Readers in other countries may subscribe for an annual subscription at the rate of $135.00.
FAX form to (317) 816-8788 or for IMMEDIATE service, simply subscribe/renew at
www.myamericanpharmaceuticalreview.com
I want to receive/renew American Pharmaceutical Review FREE YES No
Which version of American Pharmaceutical Review would you like to receive? Print Digital (include your email address below)
I want to receive APR News in Review enewletters YES No
Signature ____________________________________________________________________________ Date __________________
Name ________________________________________________________ Job Title _______________________________________
Company _________________________________________________________________________________________________________
Address __________________________________________________________________________________________________________
City ______________________________________________________ State ____________ Zip _______________________________
Phone _______________________ Fax _________________________ Email ______________________________________________
BW1X1B
subscription
& RENEWAL

82 |

|

January/February 2011
EXCIPIEnts

Irwin B. Silverstein, Ph.D.


Vice President and Chief Operating Ofcer, IPEA
email: irwin@ibsquality.com
Selling the Audit
Observation to Enhance
Conformance
Introduction
Audits are typically conducted to verify conformance to requirements
and to identify gaps between current practices and desired outcomes.
GMP audits are specifcally conducted to assess regulatory compliance
while quality audits identify opportunities to drive down cost and
improve quality while maintaining regulatory compliance and
customer satisfaction. It is generally easy to convince the auditee to
implement cost improvement opportunities identifed through quality
audit since the impact on competitiveness, and thus their livelihood, is
readily apparent. However when the GMP audit identifes a gap against
regulation or desired outcome, or compendial specifcations where
applicable, there is often the perception that the improvement does
not add value while raising costs and thus the improvement is often
considered bureaucratic.
An apt comparison can be made with driving laws. When new drivers
are informed of the rules that drivers are to follow, compliance is
based upon passing the written and then the driving test followed
by the fnancial consequences of the driver getting a ticket. However,
when compliance is sold on the personal impact to driver safety, since
compliance equates with avoiding an accident, following the rules of
the road is expected to rise to a higher level. We see this implemented
in public service advertising intended to link the afect of drinking to
unsafe driving and more recently, texting to accidents.
In this article, I will demonstrate the value of establishing the linkage
between failure to comply with regulation or specifcation and the
potential impact to patient safety. The premise is that people would
not want to knowingly jeopardize the safety of pharmaceuticals
dispensed to themselves, family members, or acquaintances let alone
to the general public.
Excipient GMPs are a guideline issued by the International
Pharmaceutical Excipients Council jointly with the Pharmaceutical
Quality Group. As such excipient GMPs are not regulation. The
interpretation of conformance expectations are those of the author
based upon his participation in the development of the initial guide in
1995 through its revisions to date.
In GMP audits of all too many excipient manufacturers quality control
laboratories, I have observed insufcient documentation of test
details. This is manifest in two aspects; frst is the failure to maintain a
permanent record of measurements particularly scale weights (gross
and tare), and the second involving recording critical aspects of test
sample preparation, e.g. time and temperature. While documentation
of such details is not an explicit requirement for excipients, it is a
practice taught in post-graduate science education. If you consider
the impact the lack of such details has on the operation of the Quality
Go for RetaLac

.
With MEGGLEs new RetaLac

, a co-processed excipient, consisting of 50% Lactose


Monohydrate and 50% HPMC you can compress HPMC directly with no problem
at all. Thanks to its good flow properties and good wettability, sustained release
tablets can be produced much more quickly, easily and efficiently (both for DC
and wet granulation). Whats more, RetaLac

gives you maximum flexibility for


your formulations. It enables the proportion of active ingredients to be as high as
60 %, and their release can be modified by adding further carriers.
RetaLac

, the worlds first Lactose-HPMC coprocessed excipient for direct com-


pression, with good flow properties, good wettability and high flexibility.
W
O
R
LD
F
IR
S
T
!
MEGGLE USA Inc.
50 Main Street, 10
th
Floor
White Plains NY 10606, USA
Phone: (914) 682 6891
Fax: (914) 682 6896
E-mail: meggle@usa.com
www.meggle-pharma.com
Mutchler Inc. Pharmaceutical Ingredients
Harrington Park, NJ , USA
Phone: 800 630 3440
Fax: (201) 768 9960
E-mail: Glenn.Mutchler@mutchlerchem.com
www.mutchlerchem.com
Meggle_Anzeigen_206x276.indd 5 27.01.11 09:37

84 |

|

January/February 2011
EXCIPIEnts

Control Laboratory, you quickly realize it greatly inhibits identifcation


of mistakes and the investigation of out-of-specifcation test results.
In order to justify retest or resample, it is benefcial to be able to
document that the original test was improperly performed based
on review of the test record. Otherwise the original result cannot be
readily negated and the incident would have to be more thoroughly
investigated as an out of specifcation test result.
While this argument would seem sufcient to motivate better
documentation in the laboratory, nothing illustrates the importance of
recordkeeping as an example. During a recent audit, the record for the
preparation of a laboratory reagent solution was reviewed. The laboratory
notebook merely listed the title of the reagent, the date, the preparer, and
the quantity of water used. What was lacking was the weight of reagent
used. Unfortunately, the title of the reagent solution recorded in the
notebook indicated a 20% solution was prepared whereas the intent was
to prepare a 15% solution. The investigation was limited to confrmation
through testing that in fact the reagent was a 15% solution. Clearly it
would have been far simpler to verify the quantity of reagent mixed with
water would yield the intended 15% solution.
This example also illustrates other failures in the laboratory including
a failure to perform an independent second check of the test record.
However, without the additional test data being recorded, a second
check would probably not have identifed the discrepancy. Had the
solution in fact been improperly prepared, there was a potential to
release excipient unknowingly with an elevated impurity.
Laboratory records should not only record the weights and volume of
materials used but also important details that demonstrate the test
was performed in accordance with the proscribed test method. If such
conditions as temperature, time, and the use of specifed laboratory
equipment are important to assuring proper sample preparation, than
those details should also be recorded.
The next example in selling the audit observation involved the
requirement to provide adequate toilet facilities. During the
inspection of a bulk loading station, a portable toilet was observed.
Upon questioning the escort, it was established that this facility was
provided for use of the loader. This operation has a sole loader each
shift and the loading station is stafed 24/7 365 days per year. Activity
at the loading station was such that the loader was unable to leave for
the time required to use the nearest available rest room.
A portable toilet provides a sanitary facility to relieve ones self but
generally does not provide for hand washing. When the lack of washing
facilities was noted, the host rationalized that the soiled hands of the
loader would not present a contamination risk to the excipient since
the hands of the loader do not come into contact with the excipient.
The auditor noted that the loader is required to take a dip sample from
the truck or railcar into a glass bottle secured in a metal holder. The
escort responded that they are required to wear gloves as a safety
requirement during all loading operations and therefore there was
no contamination risk. The auditor then pointed out that the loader
would have an easier time putting the sample bottle into the holder
without wearing bulky gloves and the escort agreed that this was
likely the case. It is important to always remember that employees will
take the easiest path. Thus it was established that the lack of a hand
washing facility presented a risk of contamination to the excipient
during bulk loading.
The next area where selling the observation was benefcial to getting
a commitment to improve the situation was in packaging. All lighting
fxtures are expected to be protected against accidental breakage
of the bulb. All light bulbs present a contamination risk due to the
fne shards of glass their breakage can introduce into the material.
Fluorescent bulbs present the added risk from the mercury they
contain. Therefore it is prudent to protect bulbs from being struck
or from falling from the fxture and shattering; particularly where
excipient is packaged.
Unprotected fuorescent fxtures were in use in the area where
excipient was being packaged into bags. The fxtures were located
roughly 7 feet above the foor and about 5 feet from the feed nozzle
at the bag packaging facility. The auditor noted the contamination risk
should a bulb break and the escort rightly observed that neither glass
nor mercury would likely get into the bag. However, the regulatory
expectation is that the bulbs of such fxtures should be protected.
It was observed during packaging that the excipient bags were stacked
below the subject lighting fxture. The auditor presented the argument
that should a bulb break, the glass and mercury would fall onto the
bags of packaged excipient. While the fne shards of glass would not
likely penetrate the bag, it would be virtually impossible to remove all
glass from the afected bags onto which the glass shards fell. It was
pointed out that excipient packaged in bags is often introduced into
the pharmaceutical manufacturing process by slitting the bag open
and shaking out the contents. It was thus apparent that glass on the
outside of the bag could very well be introduced into the process.
Therefore the potential for glass contamination in the pharmaceutical
product was evident and the manufacturer protected the light fxture.
It seems that many excipient manufacturers are reluctant to audit their
suppliers. Yet oftentimes excipient manufacturers accept incoming
raw materials solely on the basis of COA and some form of identity
confrmation. This is acceptable practice as long as there is a sound
basis for reliance on the suppliers test data as reported on the COA.
Excuses for not visiting the supplier include:
The facility is dedicated to the manufacture of the material.
The material is a commodity.
Ive used their material for quite some time without a problem.
I dont know what quality standard to audit against.
The last excuse is the simplest issue to address. The supplier audit
should be conducted against ISO 9001 since conformance to
this quality system requirement is the expectation of many non-
pharmaceutical customers.
The remaining excuses would all seem reasonable but a visit to
the supplier provides several benefts to the customer-supplier
relationship. First it personalizes the relationship between the excipient
manufacturer and their supplier. Second it educates the supplier to
the excipient application for their material and by informing them
the material is used to make a pharmaceutical ingredient; it should
motivate the supplier to maintain their conformance to specifcation.
January/February 2011 |

| 85

EXCIPIEnts
Finally, it ofers an opportunity to assure the supplier is aware of the importance in notifying the
excipient manufacturer of changes that may alter the composition or performance of the material
in the customer application.
The fnal example of selling the audit fnding deals with change control; referred to in the chemical
industry as Management of Change (MOC). Excipient manufacturers who are part of the chemical
industry generally operate under MOC due to the importance of safe operation of the facility. Any
change that can potentially afect safety in the plant is carefully reviewed under MOC before being
implemented. This facilitates addressing the issue of change control for excipient manufacture
since adding a quality review to MOC assures the impact of change to the excipient is considered.
While MOCs are carefully reviewed to assure the change is safe to implement, even where there
is a quality review of the process change for its potential to afect the quality of the excipient;
sometimes the assessment is not sufciently rigorous.
The objective in selling the audit fnding is to change the paradigm of the impact assessment of
the change on excipient quality. A site installed new equipment to afect temperature control.
While the new equipment was not a replacement in kind, an assessment of the new equipment
would conclude that it was unlikely to impact the quality of the excipient. To address the remote
possibility of product contamination, the MOC required running a fxed quantity of distilled
product through the new equipment and collecting this material for reprocessing. The reprocessed
material was to be distilled and sold.
While distillation is a good purifcation technique, the site never used distillation to assure the
removal of trace impurities left over from metal fabrication. The new equipment might contain
residues of process materials such as oils, used in its manufacture. The presence of such material
in the product processed through the new equipment to demonstrate equipment performance
and to assure the equipment was clean, can present a contamination risk. Excipient that contains
such residue which is subsequently distilled might contain trace quantities of a new impurity or
impurities. The discussion focused on the potential for this admittedly minimal risk to excipient
quality until it was noted that the company does not to reprocess or rework any excipient grade
material that has been rejected by the customer for any reason. This policy is attributable to the
small risk of introducing impurities to the excipient from customers sampling the excipient or
other activities by the customer. Once the site was reminded of this policy on customer returns, it
was apparent to all that the plan to reprocess the purge from commissioning activities conficted
with the company policy on reprocessing material returned from the customer. This intellectual
exercise has sensitized the site to a more rigorous assessment of MOC on excipient quality.
There are times when selling the observation is not apparent. The expectation is for all processing lines
to be labeled. However, the auditor is often challenged to identify the beneft to the manufacturer for
labeling the lines when the facility is dedicated to the manufacture of the excipient. Tracing the line to
the source or destination unambiguously establishes the identity of the material container therein. The
auditor is still searching for a tangible beneft where lines are fxed!
Selling the audit observation to the auditee is much more challenging than citing chapter
and verse from the regulation or guideline. It often requires quick thinking on the part of the
auditor but it can result in a paradigm shift in thinking at the audited facility. This intellectual
exercise will continue to be the best means to convince the auditee to change their practices
for improved conformance to excipient GMP until there are regulatory examples, i.e. FDA 483s
or Warning Letters, to cite.
Author Biography
Irwin Silverstein is a consultant specializing in quality assurance and regulatory compliance for
pharmaceutical excipient ingredients. He has worked since 1991 with the International Pharmaceutical
Excipients Council (IPEC) developing guidance documents for excipient GMP compliance. He has been
the VP and Chief Operating Ofcer of International Pharmaceutical Excipients Auditing Inc (IPEA) since
2001 where he was instrumental in developing the ANSI accredited IPEA Excipient GMP Conformance
Certifcation program.

86 |

|

January/February 2011
Our company is a global leader in mass spectrometry with a broad range of
innovative instrument systems, software and services used to discover new drugs,
advance medical science and protect the food supply and the environment.
AB SCIEX solutions, including the AB SCIEX TripleTOFTM 5600 System for the
fastest and most sensitive high-resolution mass spectrometer for qualitative and
quantitative analysis, combine the highest performance with the highest reliability
to enable scientists to fuel scientifc discovery and deliver results with confdence.
For more information, go to our website.
Booth # 3535
www.absciex.com
We are a leading manufacturer and supplier of specialty and high purity chemicals
available in quantities for research or production. The Alfa Aesar Catalog includes
more than 30,000 products and over 3,000 new items. In addition, the catalog also
includes a full line of Platinum Labware, Spectrofux alkali borate analytical fuxes
and the Specpure brand of analytical standards.
Booth # 4945
www.alfa.com
The Bruker name has become synonymous with the excellence, innovation, and
quality that characterizes our comprehensive range of scientifc instrumentation.
Our solutions encompass a wide number of analytical techniques ranging from
magnetic resonance to mass spectrometry, to optical and X-ray spectroscopy.
These market and technology leading products are driving and facilitating many
key application areas such as life science research, pharmaceutical analysis, applied
analytical chemistry applications, materials research and nanotechnology, clinical
research, molecular diagnostics, and homeland defense. Bruker Innovation with
Integrity!
Booth #2561
www.bruker.com
Our market leading IC systems redefned IC with RFIC, suppression, and online
capabilities and range from basic to the worlds most advanced capillary IC. Our
new UHPLC+ focused improvements make all UltiMate 3000 systems UHPLC
capable, including the high performance RSLC, RSLCnano, BioLC, semiprep and
standard. Our Chromeleon software turns samples to results fast. Our advanced
array of IC and LC column chemistries deliver unrivaled separations. Sample prep
solutions, Accelerated Solvent Extraction (ASE) systems and new AutoTrace 280
SPE.
Booth # 2861
www.dionex.com
Distek presents the ActiPix SDI300 dissolution imaging system with the unique
capability of quantitative imaging of the liquid/surface interface for a diverse range
of substances. Distek will also show their bathless and bath based Dissolution
Systems along with a variety of products for automation including; Evolution 4300
autosampler and Opt Diss In-situ UV Fiber Optics. Visit Pittcon Booth #1960 to see
the NEW products and be entered to win an iPod touch
Booth # 1960
www.distekinc.com
A global organization continuing their focus on technology strategies
encompassing a wide array of Laboratory and Scientifc instruments. Exhibiting
a product line that covers particle sizing and Zeta Potential-analyzers using both
dynamic and static light scattering, digital image analysis, optical microscopy
and acoustic attenuation technology. Highest performance in spectroscopic
instrumentation: Raman/PL microscopes with rapid imaging; spectrofuorometers;
EDXRF microscopes; ICP; C/S, O/N & H elemental analyzers; InGaAs arrays, OEM
miniature spectrometers & Raman systems & gratings. HORIBA remains committed
to global environmental conservation.
Booth # 1922, 1923
www.horiba.com
The following pages list profle and booth information for advertisers that will be exhibiting at the 2011 Pittcon Conference & Expo.

January/February 2011 |

| 87
A Rockwell Collins Company, Kaiser is recognized as a world leader in the design and
production of Raman analyzers and components for in situ Raman spectroscopy.
Kaisers suite of analyzers includes instruments for microscopy & imaging, reaction
monitoring, gas-phase Raman, solids sampling, and transmission Raman. Raman
analyzer installation locations include R&D, Pilot plant, manufacturing, and QA/QC.
Application areas for RamanRxn Systems analyzers include the pharmaceutical,
biotech, semiconductor, nanotechnology, petrochemical, polymer, and specialty
chemical areas. Kaiser ofers a range of Raman probes and optics to meet your
sampling needs.
Booth # 1648
www.kosi.com
Our company has become synonymous with expertise in weighing and analysis
instrumentation for laboratories. The laboratory division manufactures and markets
a full range of precision products including balances, pipettes, titration equipment,
thermal analysis instrumentation, density & refractive index determination
equipment, moisture analyzers, and laboratory automation systems. METTLER
TOLEDO products are fully supported by factory-trained service representatives
who perform calibration, qualifcation, and validation services.
Booth # 2726, 2727
www.mt.com
Thermal analysis, calorimetry, thermal properties, & contract testing services; DSC,
DTA, TGA, STA (Simultaneous DSC/DTA-TGA) from cryogenic to +2400C, evolved
gas analysis by coupled FTIR & MS featuring a new TGA-GC-MS system, adiabatic
reaction calorimeters (ARC & APTAC) to measure thermal & pressure properties
of exothermic chemical reactions, new MMC 274 tabletop reaction calorimeter,
thermal conductivity, thermal difusivity by laser fash & xenon fash to +2800C,
DMA, TMA, DEA for in-situ thermoset cure monitoring, & more.
Booth # 3126
www.netzsch-grinding.com/pharma
Products for HPLC/UHPLC sample prep and chromatrography applications are
available from Pall Life Sciences, the leader in Filtration and Separation.
From the newly released Advance line of flter plates and centrifuge flters, to the
industry leading Acrodisc PSF syringe flter, Pall continues to provide solutions that
improve your processes and results. Stop by our booth #2353 to see a variety of
products designed specifcally for purifcaiton, detection, sample prep and quality
control. For more information, visit Pall Life Sciences at www.pall.com/lab.
Booth # 2353
www.pall.com/biopharm
Cutting-edge technology. Ultimate commitment. PANalytical designs, develops,
and supplies X-ray analytical instrumentation and software solutions for materials
characterization. Whether in the drive for comprehensive R&D solutions or superior
quality control, PANalyticals X-ray difraction and X-ray fuorescence systems
deliver quality analytical results. Please visit us to see the latest technological
advancements in XRF, XRD and sample prep equipment, software, standards
and quality programs, all delivered with the application expertise for complete
solutions to your material analysis challenges.
Booth # 2261
www.panalytical.com
PSS is a major force in developing particle size analyzers for both wet/dry
applications. Nicomp DLS (0.5nm-6 microns) ofers nano sizing while AccuSizer
SPOS (0.15-400+ microns) ofers a wide dynamic size range providing high
resolution, high sensitivity accurate particle size information. The AccuSizer
FX PAT. ofers high concentration SPOS that has the sensitivity to detect small
diferences between particle size distributions. Our product line is rounded out
by high resolution image analysis and Archimedes SMR technology, an ultra-high
resolution mass sensor that weighs each particle, providing submicron counting
measurements of mass and size.
Booth # 1116
www.pssnicomp.com

88 |

|

January/February 2011
Phenomenex is a global technology leader committed to developing novel
analytical chemistry solutions that solve the separation and purifcation challenges
of researchers in industrial, clinical, government and academic laboratories. From
drug discovery and pharmaceutical development to disease diagnosis, food safety
and environmental analysis, Phenomenex chromatography solutions accelerate
science and help researchers improve global health and well-being.
Booth # 4634
www.phenomenex.com
We are a leader in providing automated dissolution testing systems, content
uniformity and assay workstations and automated physical tablet testing
instruments for the pharmaceutical, medical device, biopharmaceutical and
dietary supplement industries. New for 2011 are JT Bakers Dilut-IT media
concentrates, direct HPLC or UPLC analysis and UV Fiber Optic Dissolution. Learn
how to automate your pharmaceutical testing on our website.
Booth # 1447
www.sotax.com
Visit our exhibit and see worlds largest portfolio anywhere including analytical
instruments, reagents, laboratory consumables, equipment, and services.
Whether you need an instrument, an entire application workfow, or laboratory
workstations, think Thermo Scientifc. Youll fnd Thermo Scientifc innovation and
the latest products to help you run your laboratory at peak performance and run
your experiments from start to fnish. See the entire line up on our website.
Booth # 2835
www.thermo.com
Our company helps laboratory-dependent organizations by providing
breakthrough technologies and solutions. Pioneering a connected portfolio of
separation and analytical science, laboratory informatics and mass spectrometry,
Waters provides the tools to improve the quality of todays science and explore the
infnite possibilities of tomorrow. Waters, The Science of Whats Possible.
Booth # 1635
www.waters.com
Watson-Marlow Pumps, Tubing and Flexicon fllers are designed for pharmaceutical
processing and laboratory applications for fuid transfer, metering, dispensing, and
flling. These processes demand sterility and a high degree of precision to ensure
the end product meets the industrys strictly regulated quality standards. For
pumping or flling aseptically, nothing beats Watson-Marlow products. Ofering a
contamination-free single use fuid path, Watson-Marlows peristaltic technology
simplifes cleaning validation and enhances the integrity of high purity upstream
processes, purifcation, and fll/fnish applications.
Booth # 764
www.wmpg.com
WITec is a manufacturer of high resolution optical and scanning probe microscopy
solutions for scientifc and industrial applications. A modular product line allows
the combination of diferent microscopy techniques such as Raman, NSOM or AFM
in one single instrument for fexible analysis of optical, chemical and structural
properties of a sample.
Booth # 1420
www.witec.de
A complete solution
for your pharmaceutical
information needs.
9225 Priority Way West Drive, Suite 120, Indianapolis IN 46240
317-816-8787 | Fax 317-816-8788
American Pharmaceutical Review is a peer reviewed journal
that reaches over 30,000 subscribers within the North American
pharmaceutical, biopharmaceutical and contract pharmaceutical
market. Each monthly issue contains editorial tracks on
Bioprocessing, Manufacturing, PharmaIT, Aseptic Processing,
Parenteral Manufacturing, and PAT.
Our website, www.AmericanPharmaceuticalReview.com, provides
subscribers with an online extension of our magazine. Focusing on
content, the site contains over 600 articles searchable through
our advance keyword search capability. In addition, we feature
industry news, events, and white papers.
Subscribe for free at
www.americanpharmaceuticalreview.com
APR_PuzzleFull.indd 1 12/10/10 2:04:59 PM
New Chromogenic Endotoxin
Detection System
Associates of Cape Cod, Inc., (ACC) is proud to introduce its NEW
CHROMOGENIC ENDOTOXIN DETECTION SYSTEM incorporating the
enhanced Pyrochrome chromogenic reagent and the newto-market
Pyros Kinetix Flex Incubating Kinetic tube reader. This system ofers
our customers diverse options for conducting endotoxin testing.
ACCs enhanced Pyrochrome is a versatile, quantitative reagent
for performing kinetic or endpoint assays. The reagent features a
maximum sensitivity of 0.001 EU/mL, the option to use polynomial
regression and an economical sample to lysate volume ratio of 4:1.
Polynomial regression enables more accurate determination of
endotoxin concentrations, especially when a wide range standard
curve is used.
The Pyros Kinetix Flex reader is an optical tube reader that runs both
chromogenic and turbidimetric assays. Designed with fexibility and
efciency in mind, the Pyros Kinetix Flex reader and Pyros EQS
Software combine to provide a complete system that is 21 CFR Part
11 compliant for efcient, accurate endotoxin testing. AJ Meuse,
President and CEO of ACC stated We are very excited about the
opportunities that Pyrochrome and the Pyros Kinetix Flex reader
system is going to ofer the market. We understand that quality,
regulatory compliance and exceptional technical support are critical
components for our customers when deciding who they will trust with
their endotoxin release testing and this new system was designed to
satisfy our customers needs.
The Pyros Kinetix Flex reader is available in three confgurations: 32,
64 and 96 eight mm wells. Each well is independently timed, allowing
the operator to add more samples while a run is in progress. The unit
features precise temperature control, solid state design and utilizes
reliable Pyroclear depyrogenated test tubes, which eliminates
potential hot wells that are often found when using mircroplates.
ACC is an industry innovator and has been a leading global supplier
of endotoxin and glucan detection products and services for nearly
four decades. During this time, ACC has supplied our customers with
products and services that have helped ensure the safety of their
parenteral drugs, biological products and medical devices.
For more information on ACCs new Chromogenic Endotoxin Detection
System incorporating enhanced Pyrochrome reagent, Pyros EQS
Software and the Pyros Kinetix Flex Incubating tube reader, contact
Associates of Cape Cod, Inc., at 124 Bernard E. Saint Jean Drive, E.
Falmouth, MA 02536, 5085403444, www.acciusa.com.
Cook Pharmica adds E. Morrey
Atkinson, Ph.D., to Leadership Team
as Vice President of Research and
Development, Chief Scientifc Ofcer
E. Morrey Atkinson, Ph.D., has joined Cook Pharmica
as vice president of research and development and
chief scientifc ofcer, company ofcials announced
recently. Atkinson will be responsible for guiding
the scientifc direction of Cook Pharmica, relying
on his extensive past experience with process development of gene
therapies, vaccines, recombinant proteins and monoclonal antibodies.
Morrey comes to us with a broad background of scientifc and business
experience in both the domestic and international marketplace,
said Tedd Green, president of Cook Pharmica. His broad technical,
management and leadership experience will be a great strength to
our team and we are proud to welcome him to the Cook organization.
Atkinson has almost two decades of experience in biologics
development and has held various leadership positions in
biotechnology manufacturing and development. Most recently, he
was head of biotechnology manufacturing sciences and technology
for Eli Lilly and Company in Kinsale, Ireland.
I look forward to contributing my experience with biologics
development to this organization, Atkinson said. With the potential
to ofer the broadest range of development and production services
in the industry, Cook Pharmica has a bright future ahead, and I am
excited to join this leadership team.
Atkinson, who grew up in Indiana and Florida, graduated from Indiana
University with a bachelor of science in biology and received his
doctorate in biological sciences from Stanford University.
FOSS Releases New Features for Vision
FOSS NIRSystems, Inc. has introduced Service Pack 6 for Vision 3.50.
Vision is a software package specifcally designed for use with the
FOSS NIRSystems Near-Infrared (NIR) laboratory and process analyzers.
Vision 3.50 Service Pack 6 is a cumulative service pack and ofers
support for the latest NIR laboratory and process hardware.
The main new feature added in Service Pack 6:
The ProFoss instrument has been added to Vision, which
includes support for Self-Test, Diagnostics, Data Acquisition,
and Routine Analysis. ProFoss is a diode-array instrument and
the latest addition to the product line of FOSS NIRSystems.

90 |

|

January/February 2011
INDuSTRy NEwS

What are the reasons (medical, social,


environmental) in favor of needle Free PFs?
Today the statistics show that by far, the most frequent accident with
risk of blood contamination in the hospital environment is the needle
stick (74%). This is a real critical issue for the health workers (especially
the nurses) and the hospital administration that needs to report these
accidents and provide insurance coverage for the risks it involves.
Why is it more important for life saving
drugs in r2u forma?
When time is of essence, a R2U product will limit at the utmost the time
it takes to treat a patient, but also it eliminates completely any sources
of accidents and medication errors, which happens today still too often.
The French Health authorities reported in 2005 that there could be
as many as 190 000 evitable severe adverse efect events each year.
These severe events are susceptible to cause death, induce a handicap
AGUETTANT Q&A
Danielle Labreche
Director of Business Development and Innovation
Laboratoire AGUETTANT

IntErvIEW

January/February 2011 |

| 91
or prolong the hospitalization. Data from other countries confrms the
size of the issue.
The French Health Authority (AFSSAPS), published in 2009, a report on the
Medical Errors, based on 4 years of operations of their Guichet des Erreurs
Mdicamenteuses, an Adverse efect
events reporting entity. According to
this study, the most common errors
are on: drug (42%), dilution (28%);
dose (7%), and route of delivery
(6%). The source for such errors are
attributable to look alike packaging
(39%), medical procedure errors
(27%), use error (12%), and missing
information (10%).
If more secure, why are PFs not deployed to
all drugs where applicable yet?
A lot of critical care drugs have been ofered for many years in
historical primary packaging: the glass vials or glass ampoule. Years
of competition on these products has driven the market to very low
prices per unit. Now that the PFS technology exists, but at a higher cost
than vial or ampoules, how do we convince the hospital pharmacists
to pay more for security? We have to look at time saved for the staf,
reduction of medical errors, and let us not under estimate the reduction
of wasted drugs and supplies.
Nevertheless, today the PFS market is mostly addressing higher priced
drugs, new chemical entities, vaccines and new drug formulation. But
some smaller size companies have accepted to live up to the challenge.
What will it take?
The industrial supply capacity of pre-fllable syringes is still under the
market demand.
The PFS growth expectations are still at double digit growth per year,
and so the available capacity focuses on the higher valued markets.
But some smaller pharmaceutical companies, such as Aguettant, are
looking again at the critical care market, and have achieved to impose
their PFS in France on the basis of additional security and less waste.
But it took 5 years of R&D investment.
Are there any benefts of plastic over glass?
Glass is a fragile material, and heavier than plastic, it ofers less
ergonomy especially when we are talking of 5 ml, 10ml or 50ml PFS.
Also, with plastic, there are more possibilities to propose creative and
cost efective designs, for example:
Luer lock connection design, Opening system with tamper evidence
and back stopper of the plunger rod.
Types of Errors*
Total In %
Error of drug 310 42
Error of dilution 205 28
Error of dose 54 7
Error of route delivery 41 6
Other 104 17
Total 714 100
*AFSSAPS, Medical Errors Reporting
Ofce, June 2009, France
Figure 1. Nature of Exposure
Figure 2. Types of Personnel Concerned

92 |

|

January/February 2011
InDustry nEWs

If you are a current user of the FOSS Vision software, please contact us
for more information and/or a complete list of new features appearing
in version 3.50, Service Pack 6.
The FOSS Vision software is 21 CFR Part 11 compliant and supports
PAT through numerous process analysis options and process
communication capabilities. The software has an extensive security
system with multiple access levels, secure data archiving and report
generation as well as database and spreadsheet compatibility. Vision
comes with a user-friendly electronic manual with tutorials and data for
self taught hands-on method development and software operation.
scilogs second single-use Pre-
Calibrated sensor Patent Issued
SciLog, Inc. announces the issuance of the second of two single-
use sensor patents, US 7,788,047 and US 7,857,506 Disposable,
Pre-Calibrated, Pre-Validated Sensors for Use in Bio-Processing
Applications. The frst patent was issued in September 2010.
This patented technology addresses the challenges of the Process
Analytical Technology (PAT) initiative as it applies to single-use sensors
in downstream bio-processing commented Karl G. Schick, Ph.D., VP of
R&D at SciLog Inc.
SciLogs single-use, pre-calibrated sensors:
Provide real-time analytical data.
Enable pre-sterilized, closed-loop processing environments.
When used inCan be integrated into single-use, gamma-
irradiated fuid pathways (SciLog US patents 6,712,963 and
US 7,052,603), enable automation and automated data
acquisition for single-use platforms.
SciLog manufactures disposable, pre-calibrated, single-use conductivity
(SciCon), pressure (SciPres) and temperature (SciTemp) sensors. , see
www.scilog.com www.scilog.com/sensors. The sensors are designed
with process scalability in mind. They are available in, fve diferent fuid
connection sizes, ranging from Luer (Laboratory) to 1.0 TC (GMP bio-
processing), are available in each of the sensor families. Each sensor
is NIST traceable by sensor ID and comes with a calibration certifcate
and appropriate compliance statements. Sensor ID and associated
calibration data are stored in a gamma-stable memory residing within
the sensor. The stored calibration data is stable for over two years. For
more information, go to http://www.scilog.com/sensors.
These patents expand SciLogs has a signifcant patent position in
downstream, single-use technology. In addition to the newly issued
sensor patents, SciLog has received two prior patents (US Patent
6,712,963 and US Patent 7,052,603) that deal with Single-use Manifolds
for Automated, Aseptic Transfer of Solutions in Bio-Processing
Applications. SciLog has also received US Patent 7,410,587 for Liquid
Handling for Filtration and Liquid Chromatography. Specifcally,
these patents address the challenges and provide solutions relevant
to downstream, single-use purifcation by tangential fow fltration
(TFF), preparative chromatography and normal fow fltration (NFF). In
addition, SciLog received US 7,410,587 Liquid Handling for Filtration
and Liquid Chromatography.
SciLog ofers licensing arrangements of its single-use technology to
interested parties.
For further information contact:
Juliette Schick,
President, SciLog, Inc.
Ph: 608.824.0500
zymark tPW3 tablet Processing
Workstations and APW3 Active
Pharmaceutical Ingredient
Workstation
The Zymark TPW3 and APW3 automates sample preparation and
analysis for tablets, capsules, blends, creams, lotions, medical devices
and suspensions for content uniformity and stability assay testing
required in the Pharmaceutical Formulation and Quality Control
Labs. Our highly productive workstations not only increase testing
throughput and lab productivity, they also improve the quality of
analysis by minimizing operator error and eliminating variability
compared with labor intensive manual methods. If you are looking
for ways to boost your pharmaceutical labs productivity through
automation, SOTAX has the solution and experience for successful
implementation. From method development and validation to
standard operation procedures and transfer expertise, SOTAX is your
solution for pharmaceutical testing.
SOTAX Corporation
68A Elm Street
Hopkinton, MA 01748 USA
www.sotax.com
Sales Inquiries: 1-888-SOTAXUS
Email Inquiries: sotaxusa@sotax.com
Pittcon 2011 Booth #1447

January/February 2011 |

| 93

InDustry nEWs
rapid micro Biosystems Partners
with life technologies Corporation
on Automated microbial Detection
growth Direct system
Businesses team to accelerate microbial detection
and identifcation
Rapid Micro Biosystems, a leading provider of automated, non-
destructive, rapid microbial detection, announced a sales and marketing
agreement with Life Technologies Corporation, a provider of innovative
life science solutions. For quality control microbiology customers, the
agreement combines best in class automated microbial detection and
enumeration with gold-standard microbial identifcation.
The agreement leverages the strengths of both companies, benefting
customers who struggle daily with time consuming, manual processes,
and product safety testing where accuracy and time to results are
critical. The goal of the agreement is to maximize industry adoption
of the complementary technologies. The Growth Direct System
from Rapid Micro Biosystems enables rapid microbial detection,
and the MicroSEQ Rapid Microbial Identifcation System from Life
Technologies facilitates accurate bacterial and fungal identifcation.
We are excited with the opportunity this alliance presents to our
customers, said Steve Delity, Chief Executive Ofcer of Rapid Micro
Biosystems. Rapid Micro is the leader in automated rapid detection,
and through our collaboration with Life Technologies, the leader in
microbial identifcation, we create a compelling value proposition for
the marketplace.
The agreement with Rapid Micro Biosystems is part of our larger
commitment to address the needs of microbiologists in product quality
and safety testing, said Tony Hunt, General Manager of Pharma Analytics
at Life Technologies. We are pleased to add the Growth Direct System
for microbial detection to our overall microbiology portfolio.
For more information, visit
www.rapidmicrobio.com/automated_sample_control
AguEttAnt and PFIzEr Enter
Into a Patent licensing Agreement
for AguEttAnt self Flushing
Infusion Bag
AGUETTANT just comes to grant an exclusive licence to PFIZER on its
patented self fushing infusion bag, for PFIZERs systemic antifungal
molecules marketed in Europe.
This delivery system, invented and patented in Lyon, France, by
AGUETTANT, performs an automatic fushing of the connecting line
and perfusion bag after the drug delivery, without the intervention of
any medical staf.
This technology ofers several benefts for the patient and the medical
staf by:
enabling accurate delivery of the prescribed dose, without
the drug loss that is traditionally found trapped in the
connecting line;
reducing the risks of nosocomial infections via reduced
manipulations; and eliminating waste linked to the manual
preparation of the fushing;
saving time for the medical staf.
With this innovative delivery system device, both PFIZER and
AGUETTANT are striving to improve the safety and quality of care for
both patients and healthcare professionals.
nEtzsCh Premier launches line of
Deltavita nanoparticle mills Ideal
for Pharmaceutical Applications
A full line of mills for laboratory, clinical trial and
full-scale production
NETZSCH Premier Technologies, LLC introduced the DeltaVita line
of ultra-fne nanoparticle mills for wet grinding of batches ranging
0.05 to 2000 liters. The new line features NETZSCHs proprietary ZETA
grinding system and comprises 10 mills, designed to accommodate
the entire manufacturing process with repeatability and scalability
from testing through development to full-scale production.
Through the use of high-energy, high fow-rate, multiple-pass
grinding, NETZSCHs DeltaVita line achieves excellent repeatability
and homogeneous dispersion. In the time it takes an ordinary mill to
complete one pass, DeltaVita mills can complete as many as 10 cycles.
The ZETA grinding system ofers a single-tank process to reduce
contamination, maintain precision temperature control and provide
an easy-to-clean design.
The DeltaVita 15 through 300 systems use grinding media from
0.05 mm to 0.2 mm for consistent particle reductions to below 200
nanometers. They can be fitted with variable grinding chamber
sizes, making them ideal for feasibility studies where the smallest
batch sizes are needed to achieve significant test results in a
short period of time. Grinding zone parts are manufactured with
stabilized Zirconia/Yttrium, a high-strength ceramic, for metal-and
contamination-free grinding.
For clinical trial phase production, the DeltaVita 600 can produce
batch sizes of one to 6 liters. It features interchangeable agitating
systems, optional explosion-proof design and optional PLC control.
The DeltaVita 2000 through 60000 provide batch sizes ranging from
50 to 2000 liters.

94 |

|

January/February 2011
InDustry nEWs

All sizes of NETZSCH DeltaVita mills feature:


Wetted parts designed and manufactured according to the
latest GMP standards;
Material, production and calibration certifcates included;
Optional cleaning in place (CIP) and sterilization in place
(SIP) capability;
All indirectly product-wetted surfaces made of stainless steel;
Optional data recording and formulation management;
Operator management with password protection for
diferent levels of security;
Various materials (such as ZrO2, stainless steel 316 or nylon)
available for custom chamber design;
Splash-proof machine stand;
Comprehensive testing and qualifcation documentation; and
Training sessions and seminars available.
All NETZSCH pharmaceutical process equipment, including the
DeltaVita series, meets the high demands of the pharmaceutical
industry. NETZSCH has a dedicated team for pharmaceutical
applications experienced in the industry-specifc requirements for
testing, validation, and documentation including FAT, IQ, OQ, URS, FRA
and DDS.
For more information, visit http://www.netzsch-grinding.com or call
484-879-2020.
Crystal Pharmatech
Crystal Pharmatech Co., Ltd is currently the only high-quality, dedicated
solid state research company based in China. Our researchers have
extensive experience in top pharmaceutical companies with API and
drug product development. Whether you are a top-tier innovator
company or a pre-emerging biotech, Crystal Pharmatech can handle
all of your solid-state research and development needs. We ofer a full
range of services from being the solid state characterization arm of
your company to consultation or training on a specifc issue.
Solid state research and development support is critical to speeding
up drug development, lowering costs, and providing the highest
quality product to innovator pharmaceutical companies. explains
Alex M. Chen, CEO of Crystal Pharmatech. China is currently the top
choice for pharmaceutical outsourcing owing to the strong support
from government and vast talent pool of relatively low cost labor.
It is inevitable that China-based companies focused on providing
solid state R&D solutions will be an integral component to the
outsourcing package.
Besides providing contracted research services, Crystal Pharmatech
Co., Ltd, will also be developing new technologies and proprietary
devices that will improve efciency and drive down development
costs for all pharmaceutical companies.
For more information, visit: http://www.crystalpharmatech.com/
Particle sizing systems
When Size Really Matters, You can Count on
Particle Sizing Systems
Particle Sizing Systems announces that it is updating its look with a
new logo. Our company has evolved over the past 33 years as a major
force in the particle sizing industry. We have had a few logos along the
way. However, the one thing that remains constant with PSS is that we
continue to patent and manufacture particle sizing instrumentation
that sets us apart from the others that are available in the market
place today. We have dedicated and continue to dedicate ourselves
to developing leading edge technology and producing innovative
instruments that ofer unique, powerful capabilities for laboratory and
production environments. We will continue working to serve, support
and provide new innovations in the feld of particle sizing.
Please visit our new website to see all of the particle sizing instruments
that we have to ofer.
www.pssnicomp.com
B&W tek, Inc Announces Improved
Quest series miniature Fiber optic
spectrometers
B&W Tek, Inc., an advanced instrumentation company producing
optical spectroscopy and laser systems announces the updated
Quest series of miniature fber optic spectrometers. The Quest
series of high performance miniature fber optic spectrometers now
features an ultralow thermal drift spectrum of ~19 counts/C typical
and a faster readout speed of >2.0MHz. As an added bonus, the
Quest series is now available with optional RS232 communication
interface for convenient integration into larger systems.
The fast read out speed of the new Quest series spectrometers
makes them ideal for high speed applications such as LED binning and
sorting, says Robert Chimenti, Marketing Manager for B&W Tek, Inc.
We feel that this will strengthen our position as a leader in the LED
characterization market.
The Quest series employs both a traditional crossed Czerny Turner
spectrograph (Quest X) as well as an unfolded Czerny Turner
spectrograph (Quest U) which minimizes stray light in the UV region.
The series is equipped with 2048 elements linear CCD array, built-in
16-bit digitizer, and an externally synchronized trigger.
For more information on the Quest series, please visit http://www.
bwtek.com/product/spectrometer/.

ClAssIFIED ADvErtIsEmEnts

January/February 2011 |

| 95

www.acciusa.com
800.LAL.TEST (525.8378)
custservice@acciusa.com
If You Dont, Youre Probably
Paying Too Much.
DO YOU KNOW YOUR ENDOTOXI N
COST PER TEST?
Contact us by calling
800-724-4158
www.advancedscientifcs.com
single use systems
Achieving Faster Time
to First in Man
Capsugel R&D benchtop machines.
www.capsugel.com
APRBnr_17373_v1_2-19-09.indd 1 2/19/09 4:31:14 PM
PAT-aligned endotoxin testing
www.laboftomorrow.com
Rapid Testing
Solutions
www.lonza.com/rts
Get results now
RTS_classified.indd 1 8/24/10 3:42 PM
C
M
Y
CM
MY
CY
CMY
K
11-4237_KSDbiomall82.5x25.7_DM.pdf 24/01/11 13:52:59
Call +1 866-PATHEON (+1 866-728-4366)
or email doingbusiness@patheon.com
Why limit your compound to just one
bioavailability enhancement technology?
Congurable solutions.
Flexible, ready-to-connect
unit operations.
www.sartorius-stedim.com/exact
turning science into solutions
Ad_FlexAct_3,25x1,22inch.indd 1 15.03.10 08:24
Congurable solutions.
Flexible, ready-to-connect
unit operations.
www.sartorius-stedim.com/exact
turning science into solutions
Ad_FlexAct_3,25x1,22inch.indd 1 15.03.10 08:24

96 |

|

January/February 2011
ADvErtIsErs InDEX

Company Page # Phone # Web Address


AAPS IBC 703-243-2800 www.aapspharmaceutica.com
AB Sciex 19 877-740-2129 www.absciex.com
Accugenix, Inc. 54 302-292-8888 www.accugenix.com
Advanced Scientifcs, Inc. 63 marketing2@advancedscientifcs.com www.advancedscientifcs.com
Aguettant 77 +33 (0)4 78 61 51 41 www.aguettant.com
Alfa Aesar 85 978-521-6300 www.alfa.com
Associates of Cape Cod 6,53 888-395-2221 www.acciusa.com
Blake Hotel 80 888-664-6835 blakehotelnc.com
Bruker 25 888-4BRUKER www.bruker.com
Cambridge Healthtech Institute 65 888-999-6288 www.healthtech.com
Capsugel 1 888-783-6361 www.capsugel.com
Catalent Pharma Solutions 67 866-720-3148 www.catalent.com
Charles River IFC askcharlesriver@crl.com www.criver.com
Crystal Pharmatech 69 855-546-4986 www.crystalpharmatech.com
Dionex 15 408-737-0700 www.dionex.com/chromeleon7
Distek, Inc. 45 888-234-7835 www.distekinc.com
Enwave Optronics 40 949-955-0258 www.enwaveopt.com
GE Healthcare Life Sciences 60 800-526-3593 www.gelifesciences.com
Horiba Scientifc 41 732-494-8660 www.horiba.com/scientifc
ICDD - International Centre for Difraction Data 27 610-325-9814 www.icdd.com
Intercontinental Hotel 57 800.424.6835 www.intercontinental.com/baltimore
Interphex 11 203.840.5447 www.interphex.com
Kaiser Optical Systems, Inc. 37 734-665-8083 www.kosi.com
Life Technologies 64 760-603-7200 www.lifetechnologies.com
Lonza 51 800-638-8174 www.lonza.com/moda
Meggle Group 83 914-682-6891 www.meggle-pharma.com
Mettler Toledo, Inc. 43 800-METTLER www.mt.com
Netzsch 31 484-879-2020 www.netzsch-grinding.com/pharma
Pall Life Sciences 59 biopharm@pall.com www.pall.com/biopharm
PANalytical 23 +31 (0)546 534444 www.panalytical.com
Particle Sizing Systems 33 727-846-0866 www.pssnicomp.com
Patheon 71 919-226-3200 www.patheon.com
Parenteral Drug Association (PDA) 3 301-656-5900 www.pda.org
Perfex 5 800-848-8483 www.perfexonline.com
Phenomenex Inc. 13 310-212-0555 & info@phenomenex.com www.phenomenex.com
Rapid Micro BioSystems 55 781-271-1444 www.rapidmicrobio.com
Sartorius Stedim North America 9 800-368-7178 www.sartorius-stedim.com
Sotax 47 1-888-SOTAXUS www.sotax.com
Thermo Fisher Scientifc OBC 978-642-1132 www.thermo.com
ToxExpo 29 703-438-3115 www.toxexpo.com
Waters Corporation 17 800-252-4752 www.waters.com
Watson-Marlow Pumps Group 61 800-282-8823 www.wmpg.com
West Pharmaceuticals 75 800-345-9800 www.westpharma.com
Witec 39 865-984-4445 www.witec.de

The contact directory is for information purposes only. While every efort has been made to ensure it is accurate and complete, any errors or omissions are not the responsibility of the publisher.
Mark Your
Calendar!
For Up-To-Date Information
www.aapspharmaceutica.com/nationalbiotech
MAY 16
~
18
2011
Hilton San Francisco
Union Square
SAN FRANCISCO

CA
C
M
Y
CM
MY
CY
CMY
K

2
0
1
1

T
h
e
r
m
o

F
i
s
h
e
r

S
c
i
e
n
t
i
f
i
c

I
n
c
.


A
l
l

r
i
g
h
t
s

r
e
s
e
r
v
e
d
.
Raw material identifcation just got exciting!
Introducing an exciting new advance in raw material
identification the Thermo Scientific TruScan RM analyzer.
Our lightest, fastest and most portable handheld analyzer
utilizes the power of Raman spectroscopy to increase incoming
inspection rates saving you time and money.
Combining a state-of-the-art optical platform with enhanced
decision algorithms, the analyzer simplifies everything from
method development to routine authentication.
The TruScan

RM analyzer lets you perform identification of a


broad range of chemical compounds through sealed packaging
with more speed and reliability than ever before.
Work faster. Work smarter. Get excited!
Learn more at www.thermoscientific.com/RMID
Moving science forward
Thermo Scientific TruScan RM Analyzer
Providing GMP facilities with cost-effective
and efficient raw material identification.

Anda mungkin juga menyukai