helloalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to...

62
Introducing Nuvia cPrime media, the new hydrophobic cation exchange media that delivers unique selectivity and more purification power than ever before. With simpler method development and a wide design space, Nuvia cPrime lets you achieve a higher level of performance with a lot less compromise — so you can get to the clinic faster. HELLO NEW SELECTIVITY. (GOODBYE LIMITING COMPROMISE.) Download the application note at bio-rad.com/ad/cPrime

Upload: buinhan

Post on 18-Mar-2018

220 views

Category:

Documents


3 download

TRANSCRIPT

Page 1: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

Introducing Nuvia™ cPrime™ media, the new hydrophobic cation exchange media that delivers unique selectivity and more purification power than ever before. With simpler method development and a wide design space, Nuvia cPrime lets you achieve a higher level of performance with a lot less compromise — so you can get to the clinic faster.

HELLO

NEW SELECTIVITY.(GOODBYE LIMITING COMPROMISE.)

Download the application note at bio-rad.com/ad/cPrime

Page 2: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

With a unique balance between modes

of interaction, Nuvia cPrime can

effectively retain the full-length mAb at

pH 5.0 while allowing a 25 Kd L-chain

fragment to flow-through the column,

without any conductivity adjustments

from the previous ion exchange

purification step. Elution at pH 6.2

yields L-chain fragment and aggregate

free antibody collected under mild

conditions and with high recovery.

Achieve new selectivity without

compromise. Simplify complicated

multi-step processes that require

cumbersome conductivity and pH

adjustments by effectively employing

the versatile mixed-mode properties of

Nuvia cPrime at any suitable step in a

multi-column process.

Download the application note at www.bio-rad.com/ad/cPrime

NUVIA cPRIME MEDIA IN A THREE STEP NON-PROTEIN A mAb PURIFICATION PROCESS

12.5

10.0

7.5

5.0

1.75

1.50

1.25

1.00

0.75

0.50

0.25

0.00

1 2 3 4 5 6 7 8 9 10 16 17 18 19 20 21 22 23 24 25Fraction

AU

pH

Time, min

0.00 30.00 60.00

100% Buffer G

50%

Flowthrough EluatemAb1

Buffer Hstrip

1 N NaOHstrip

It’s time to say Hello to new selectivity while waving Goodbye to limiting compromise.

Page 3: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

The Science & Business of Biopharmaceuticals

www.biopharminternational.com

INTERNATIONAL

BioPharm

Bio

Ph

arm

Intern

atio

nal

NO

VEM

BER 2

012

B

iosim

ilars I S

tem

-Cell S

cale

-up

I An

aly

tical M

eth

od

s V

olu

me 2

5 N

um

ber 1

1

November 2012

Volume 25 Number 11

BIOSIMILARSNEW GUIDELINES SET FOR

MONOCLONAL ANTIBODIES

PEER-REVIEWED:

A SINGLE-USE FLUIDIZED

BED CENTRIFUGE SYSTEM

FOR MAMMALIAN

CELL HARVESTING

SCALE UP:

KEY CONSIDERATIONS

FOR WORKING WITH

STEM-CELL CULTURES

HOW TO:

�t�*.1307&�"/"-:5*$"-�.&5)0%�

%&7&-01.&/5�*/����45&14

t�$0//&$5�,/08-&%(&�

."/"(&.&/5�50�4611-*&3�26"-*5:

Page 4: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

DAWN HELEOS®. The most advanced multi-angle light scattering instruments for absolute macromolecular characterization.

Optilab T-rEX®. The refractometer with the greatest sensitivity and range.

ViscoStar®. The viscometer with unparalleled signal-to-noise, stable baselines and a 21st-century interface.

Eclipse. The ultimate system for the separation of macromolecules and nanoparticles in solution.

DynaPro Plate Reader II. Automated dynamic light scattering for proteins and nanoparticles in 96 or 384 or 1536 well plates, and now with an on-board camera!.

Page 5: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

The Science & Business of Biopharmaceuticals

INTERNATIONAL

BioPharm

©2012 Advanstar Communications Inc. All rights reserved. No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical including by photocopy, recording, or information storage and retrieval without permission in writing from the publisher. Authorization to photocopy items for internal/educational or personal use, or the internal/educational or personal use of specific clients is granted by Advanstar Communications Inc. for libraries and other users registered with the Copyright Clearance Center, 222 Rosewood Dr. Danvers, MA 01923, 978-750-8400 fax 978-646-8700 or visit http://www.copyright.com online. For uses beyond those listed above, please direct your written request to Permission Dept. fax 440-756-5255 or email: [email protected].

Advanstar Communications Inc. provides certain customer contact data (such as customers’ names, addresses, phone numbers, and e-mail addresses) to third parties who wish to promote relevant products, services, and other opportunities that may be of interest to you. If you do not want Advanstar Communications Inc. to make your contact information available to third parties for marketing purposes, simply call toll-free 866-529-2922 between the hours of 7:30 a.m. and 5 p.m. CST and a customer service representative will assist you in removing your name from Advanstar’s lists. Outside the U.S., please phone 218-740-6477.

BioPharm International does not verify any claims or other information appearing in any of the advertisements contained in the publication, and cannot take responsibility for any losses or other damages incurred by readers in reliance of such content.

BioPharm International welcomes unsolicited articles, manuscripts, photographs, illustrations, and other materials but cannot be held responsible for their safekeeping or return.

To subscribe, call toll-free 888-527-7008. Outside the U.S. call 218-740-6477.

EDITORIALEditorial Director Angie Drakulich [email protected]

Managing Editor Susan Haigney [email protected]

Scientific Editors Amy Ritter, PhD, [email protected] and Adeline Siew, PhD, [email protected]

Community Managers Stephanie Sutton [email protected] and Chris Allen [email protected]

Director of Content Peter Houston [email protected]

Art Director Dan Ward [email protected]

Contributing Editors Jill Wechsler, Jim Miller, Eric Langer, Anurag Rathore, Jerold Martin, and Simon Chalk Correspondents Hellen Berger (Latin & South America, [email protected]), Jane Wan (Asia, [email protected]), Sean Milmo (Europe, [email protected])

ADVERTISINGPublisher Mike Tracey [email protected]

National Sales Manager Steve Hermer [email protected]

European Sales Manager Richard Hodson [email protected]

Market Development, Classifieds, and Recruitment Tod McCloskey [email protected]

Direct List Rentals Tamara Phillips [email protected]

Reprints The YGS Group [email protected], 800.290.5460 ext 100 or +1.717.505.9701 ext 100

Vice President Sales/Group Publisher Russ Pratt [email protected]

PRODUCTION, MARKETING, CIRCULATIONProduction Manager Jesse Singer [email protected]

Audience Development Manager Nidia Augustin [email protected]

Joe Loggia, Chief Executive Officer; Tom Florio, Chief Executive Officer Fashion Group, Executive Vice-President; Tom Ehardt, Executive

Vice-President, Chief Administrative Officer; Steve Sturm, Executive Vice-President, Chief Marketing Officer; Georgiann DeCenzo, Executive

Vice-President, Healthcare, Dental & Market Development; Chris DeMoulin, Executive Vice-President, Customer Development

& President, Licensing International; Danny Phillips, Executive Vice-President, Powersports; Ron Wall, Executive Vice-President,

Pharmaceutical/Science, CBI, and Veterinary; Eric I. Lisman, Executive Vice-President, Corporate Development; Francis Heid, Vice-President,

Media Operations; Michael Bernstein, Vice-President, Legal; Nancy Nugent, Vice-President, Human Resources; J Vaughn, Vice-President,

Electronic Information Technology

EDITORIAL ADVISORY BOARDBioPharm International’s Editorial Advisory Board comprises distinguished specialists involved in the biologic manufacture of therapeutic drugs, diagnostics, and vaccines. Members serve as a sounding board for the editors and advise them on biotechnology trends, identify potential authors, and review manuscripts submitted for publication.

K. A. Ajit-Simh President, Shiba Associates

Rory Budihandojo Manager, Computer Validation Boehringer-Ingelheim

Edward G. Calamai Managing Partner Pharmaceutical Manufacturing and Compliance Associates, LLC

John Carpenter Professor, School of Pharmacy University of Colorado Health Sciences Center

Suggy S. Chrai President and CEO The Chrai Associates

Janet Rose Rea Vice President, Regulatory Affairs and Quality Poniard Pharmaceuticals

John Curling President, John Curling Consulting AB

Rebecca Devine Biotechnology Consultant

Leonard J. Goren Global Leader, Human Identity Division, GE Healthcare

Uwe Gottschalk Vice President, Purification Technologies Sartorius Stedim Biotech GmbH

Fiona M. Greer Global Director, BioPharma Services Development SGS Life Science Services

Rajesh K. Gupta Laboratory Chief, Division of Product Quality Office of Vaccines Research and Review CBER, FDA

Chris Holloway Group Director of Regulatory Affairs ERA Consulting Group

Ajaz S. Hussain VP, Biological Systems, R&D Philip Morris International

Jean F. Huxsoll Senior Director, QA Compliance Bayer Healthcare Pharmaceuticals

Barbara K. Immel President, Immel Resources, LLC

Denny Kraichely Associate Director Johnson & Johnson

Stephan O. Krause Principal Scientist, Analytical Biochemistry, MedImmune, Inc.

Steven S. Kuwahara Principal Consultant GXP BioTechnology LLC

Eric S. Langer President and Managing Partner BioPlan Associates, Inc.

Howard L. Levine President BioProcess Technology Consultants

Herb Lutz Senior Consulting Engineer Millipore Corporation

Hans-Peter Meyer VP, Innovation for Future Technologies Lonza, Ltd.

K. John Morrow President, Newport Biotech

Barbara Potts Director of QC Biology, Genentech

Tom Ransohoff Senior Consultant BioProcess Technology Consultants

Anurag Rathore Biotech CMC Consultant Faculty Member, Indian Institute of Technology

Susan J. Schniepp Vice-President Quality and Regulatory Affairs Allergy Laboratories, Inc

Tim Schofield Managing Director Arlenda, USA

Paula Shadle Principal Consultant, Shadle Consulting

Alexander F. Sito President, BioValidation

S. Joseph Tarnowski Senior Vice President, Biologics Manufacturing & Process Development Bristol-Myers Squibb

William R. Tolbert President, WR Tolbert & Associates

Michiel E. Ultee Chief Scientific Officer Laureate BioPharmaceutical Services, Inc.

Thomas J. Vanden Boom Vice President, Global Biologics R&D Hospira, Inc.

Krish Venkat Principal AnVen Research

Steven Walfish President, Statistical Outsourcing Services

Gary Walsh Associate Professor Department of Chemical and Environmental Sciences and Materials and Surface Science Institute University of Limerick, Ireland

Lloyd Wolfinbarger President and Managing Partner BioScience Consultants, LLC

Page 6: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

4 BioPharm International www.biopharminternational.com November 2012

Contents

BioPharmINTERNATIONAL

BioPharm International integrates the science and business of

biopharmaceutical research, development, and manufacturing. We provide practical,

peer-reviewed technical solutions to enable biopharmaceutical professionals

to perform their jobs more effectively.

ON THE WEBwww.biopharminternational.com

COLUMNS AND DEPARTMENTS

BioPharm International ISSN 1542-166X (print); ISSN 1939-1862 (digital) is published monthly by Advanstar Communications, Inc., 131 W. First Street, Duluth, MN 55802-2065. Subscription rates: $76 for one year in the United States and Possessions; $103 for one year in Canada and Mexico; all other countries $146 for one year. Single copies (prepaid only): $8 in the United States; $10 all other countries. Back issues, if available: $21 in the United States, $26 all other countries. Add $6.75 per order for shipping and handling. Periodicals postage paid at Duluth, MN 55806, and additional mailing offices. Postmaster Please send address changes to BioPharm International, PO Box 6128, Duluth, MN 55806-6128, USA. PUBLICATIONS MAIL AGREEMENT NO. 40612608, Return Undeliverable Canadian Addresses to: Pitney Bowes, P. O. Box 25542, London, ON N6C 6B2, CANADA. Canadian GST number: R-124213133RT001. Printed in U.S.A.

BIOSIMILARS

EU Sets Guidelines for

Biosimilar mAbs

Sean MilmoThe European Medicines Agency has added

granularity to its biosimilars approval pathway

by releasing a guideline on mAbs. 24

UPSTREAM PROCESSING

Considerations for

Scale-Up of Stem-Cell Cultures

Matthieu Egloff and Jose CastilloScaling up stem-cell cultures requires careful

consideration of the bioreactor design. 28

PEER–REVIEWED

Evaluation of Single-Use

Fluidized Bed Centrifuge

System for Mammalian

Cell Harvesting

Hsu-Feng Ko and Ravi BhatiaThis article discusses the evaluation of a

novel single-use fluidized bed centrifuge for

harvesting of antibodies. 34

ANNIVERSARY

RETROSPECTIVE

A 25-Year Retrospective on

Protein Recovery through

Membrane Filters 23

BioPharm International�JT�TFMFDUJWFMZ�BCTUSBDUFE�PS�JOEFYFE�JO��r�Biological Sciences Database (Cambridge Scientific Abstracts)�r�Biotechnology and Bioengineering Database (Cambridge Scientific Abstracts)�r�Biotechnology Citation Index (ISI/Thomson Scientific)�r�Chemical Abstracts (CAS) rŞScience Citation Index Expanded (ISI/Thomson Scientific)�r�Web of Science (ISI/Thomson Scientific)

6 From the Editor

Social media is reshaping disease prevention. Angie Drakulich

7 Global News

10 Regulatory Beat

Manufacturing and in-depth characterization provide basis for demonstrating product equivalence.Jill Wechsler

14 Perspectives on Outsourcing

Procurement organizations are rethinking sourcing for maximum efficiency and results.Gregg Brandyberry

16 Compliance Notes

Understanding supplier capability can reduce risk and cost. J. Paul Catania

20 Disposables Advisor

Are you making the best choices in single-use tubing?Jerold Martin

41 Boot Camp: Tech Guide

NIBRT’s Jayne Telford provides an overview of biopharmaceutical analytics and their accompanying qualification and validation steps.

45 Boot Camp: Business Guide

Patrick Jackson of Vindon Scientific offers key considerations for choosing an outsourced sample storage facility.

48 Bioanalytical Best Practices

A 10-step systematic approach to analytical method development and validation.Thomas Little

52 Manufacturing Best Practices

Is process-centered organization a stepping stone or a stumbling block?Simon Chalk

54 Spotlight

55 New Technology Showcase

56 Ad Index/Calendar/

Marketplace

58 Final Word

A disciplined approach to changing behavior can achieve change agility.Tracy Thurkow, Karen

Gorman, and Paula Butte

Social Media

Follow us on Twitter@BioPharmIntl

Join our BioPharmInternational Group

Special Issue: Single Use Be sure to check out our Single-Use Technologies and Facilities special supplement.

Cover: Photodisc/Thinkstock

Volume 25 Number 11 November 2012

FEATURES

Page 7: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

Cedex Bio HT – Bioprocess AnalyzerBe confident. Be assured.

United States +1 800 428 5433, ext. 14649 (toll-free)[email protected]

Canada +1 450 686 [email protected]

Europe, Middle East, Africa, and Latin America +49 621 759 [email protected]

Japan +81 3 5443 [email protected]

Asia Pacific +65 6371 [email protected]

Your Roche Custom Biotech Customer Service

cedexbio.roche.com

Roche Diagnostics GmbHSandhofer Straße 11668305 Mannheim, Germany

© 2012 Roche Diagnostics. All rights reserved.

For use in quality control/manufacturing process only.

CEDEX is a trademark of Roche.

Next generation of cell culture process control

Confidence

Speed

�� Flexible plug-and-play reagent format�� Reagent and sample identification via barcode�� High-level system robustness and reliability

resulting in benchmarking test precision

�� Parallel processing of selectable assay profiles�� Automated onboard sample dilution�� High throughput (approx. 320 tests/hour)

�� True walk-away analyzer – minimal hands-on time�� Consistent results between Cedex Bio and Cedex Bio HT Analyzers

Value

Page 8: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

6 BioPharm International www.biopharminternational.com November 2012

From the Editor

Global coordination

tactics that

incorporate online

technologies and

social media are

reshaping disease

prevention and

response.

Knowledge is Power

On Oct. 3, 2012, NPR’s Richard Knox reported on a growing trend in identi-

fying infectious diseases and new viruses. The World Health Organization

(WHO) issued an alert in September about a new SARS-related coronavirus

that affected a Qatari man who had recently visited Saudi Arabia. Another Saudi

Arabian man died in July from the same virus. In the US, the Centers for Disease

Control and Prevention (CDC) identified a new tick-borne virus, being called the

Heartland virus, in Missouri in late August. And recently in the Democratic Republic

of Congo, researchers identified a new Rhabdovirus associated with hemorrhagic

fever, based on an outbreak that started in 2009.

“The most striking thing about all three new viruses is that they were found on

the basis of just two or three human cases,” said Knox in his NPR report. “That’s a

long way from where the world was 10 years ago, when another new virus popped

up in Asia [the original SARS] and quickly went global.” In fact, the 2003 SARS virus

affected 8000 people in 26 countries before it was brought under control, according

to the WHO website.

“Today, we would have seen that information bubbling up in many different

places,” John Brownstein, PhD, of Boston’s Children’s Hospital (BCH) told Knox in

the NPR report. Brownstein helped to create a real-time website called Healthmap.

org and a related mobile app called Outbreaks Near Me. Both technologies collect and

post information on global disease outbreaks 24 hours a day, 365 days a year, with the

goal of facilitating early detection of global public health threats.

Digital disease-detection programs like Healthmap are having a huge effect on how

fast virus news can spread. Google has a population-tracking website for influenza

epidemics that monitors trends worldwide. A similar tracker, Influenzanet, has been

monitoring flu-like viruses across Europe since 2003. Canada’s Global Public Health

Intelligence Network is an Internet-based early-warning system that gathers public

health reports in real time from global media sources in six languages. And the US

CDC’s BioSense 2.0 program, which launched in 2011, tracks evolving health prob-

lems across the country to get word out to healthcare authorities and patients quickly.

Taking this a step further, President Obama enacted the first US National Strategy for

Biosurveillance over the summer. The strategy enables the Department of Defense

to track and analyze disease-related activity with an eye towards early warning and

detection. The system is based on the department’s counterterrorism work.

Another catalyst for the move towards rapid global disease identification and

response has to do with new global regulations. WHO enacted new binding

International Health Regulations in 2005 (they hadn’t been updated since 1969 and,

until 2005, only included cholera, yellow fever, and dengue) that require countries to

report certain disease outbreaks and public-health events.

And let’s not forget the power of social media platforms. At the BCH’s 2012 inter-

national conference on digital-disease detection, an entire session was devoted to

Twitter, in which John Dredze of Johns Hopkins University, among others, spoke

about using social media to mine public health information. Using algorithms,

Dredze’s team identified 1.6 million health-related tweets between April 2009 and

October 2012, and categorized them into 15 fields, such as common cold, cancer,

and depression. Courtney Corley and his team at the Pacific Northwest National

Laboratory presented research into how social media conversations can be analyzed

to identify emerging concerns about vaccine safety. In general, social media and

the Internet have opened a new world in which scientists, healthcare professionals,

academics, regulators, and patients can work together to bring light to new or grow-

ing diseases before they spread in mass numbers, thereby strengthening global public

protection in ways we never thought possible. As new viruses and infectious diseases

emerge—and they will—it’s good to know that we have these information technolo-

gies at our disposal. As the saying goes, knowledge is power. z

Angie Drakulich is the

editorial director of

BioPharm International.

Page 9: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

sidebartext_VR_subhead sidebartext_VR_

subhead sidebartext_VR_subhead

sidebartext_VR_noindent sidebartext_VR_noindent sidebartext_VR_noindent sidebartext_VR_noindent sidebartext_VR_noindent

vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_

sidebartext_VR_subhead sidebartext_VR_

subhead sidebartext_VR_subhead

sidebartext_VR_noindent sidebartext_VR_noindent sidebartext_VR_noindent sidebartext_VR_noindent sidebartext_VR_noindent

vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_sidebartext_vaccine report_

head_sans head_sans head_sanstextnoindent_sans textnoindent_sans textnoindent_sans textnoindent_sans

textnoindent_sans textnoindent_sans

text_sans text_sans text_sans text_sans text_sans text_sans text_sans text_

sans text_sans text_sans text_sans

subhead_ subhead_ subhead_

textnoindent_sans textnoindent_sans textnoindent_sans textnoindent_sans

textnoindent_sans textnoindent_sans

text_sans text_sans text_sans text_sans text_sans text_sans text_sans text_

sans text_sans text_sans text_sans text_sans text_sans

—bylinename_

head_serif head_serif head_serifAuthors: bylinebio_ bylinebio_ bylinebio_ bylinebio_ bylinebio_ bylinebio_

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

text_serif text_serif

text_serif text_serif

text_serif text_serif

text_serif text_serif

text_serif text_serif

text_serif

subhead_ subhead_

subhead_

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

textnoindent_serif

text_serif text_serif

text_serif text_serif

text_serif text_serif

text_serif text_serif

text_serif text_serif

text_serif

Discovery Pipeline

Ph

oto

cre

dit

: iS

toc

kp

ho

to/T

hin

kS

toc

k im

ag

es

Improved Delivery of RNA Vaccines

Creating an effective nucleic acid-based vaccine requires protecting the fragile nucleic acid from degradation, effective transfection of the targeted cells, and producing high enough levels of antigen to evoke a robust immune response. A group of researchers from Novartis demonstrates how to accomplish these goals using self-replicating RNA encased in a lipid nanoparticle delivery system. In a paper published online Sept. 4, 2012 in the Proceedings of the National Academy of Sciences, the group describes the delivery system, a lipid nanoparticle similar in formulation to nanoparticles used for small interfering RNA delivery. The formulation was efficient at encapsulating RNA, and produced consistently sized particles of between 79-121 nm (mean diameter). The particles protected the RNA from degradation, and were required for efficient transfection of muscle cells.

To produce the vaccine antigen, the researchers used a self-amplifying RNA based on an alphavirus genome. The RNA construct contained genes required to replicate the RNA strand, but lacked the genes for the viral structural proteins necessary to make an infectious particle. Instead, the structural genes were replaced with the genes for the vaccine’s antigen. This type of construct will replicate itself within the cell cytoplasm and does not require transport to the cell nucleus or integration into the host cell’s genome. The authors created a self-amplifying RNA vaccine against respiratory syncytial virus, and formulated it using lipid nanoparticles. In mice, the vaccine produced a robust immune response and protection against infection comparable to a vaccine made using infectious viral particles. The authors suggest that this system offers advantages over current technologies, including the ability to produce the vaccine synthetically, elimination of the risks of working with infectious particles, and avoidance of immunogenicity against a viral vector.

Source: Geall et al., Proc. Natl. Acad. Sci. online, DOI:10.1073/

pnas.1209367109, Sept. 4, 2012.

Report from TurkeyThe government of Turkey is drawing

up a program in coordination with

the pharmaceutical industry to

create ways to make the country

a regional production center for

pharmaceuticals serving Europe,

Central Asia, and the Middle East.

Turkey’s two main pharmaceutical

trade associations—Pharmaceutical

Manufacturers’ Association of Turkey (IEIS) and the Association of Research

Based Pharmaceutical Companies (AIFD)—have been supporting the

government’s initiative with the publication of reports on the future of the

Turkish drugs industry by two leading US-based consultants. AIFD published a

report, Vision 2023, by PriceWaterhouseCoopers (PwC) in September 2012 and

IEIS issued a study by the Boston Consulting Group (BCG) late last year (1, 2).

Turkey, one of the world’s fastest growing emerging economies, faces

considerable challenges in achieving its aim of becoming a major global power

in pharmaceuticals.  Within its own market, it will have to triple the average

annual growth rate and raise the annual increase of domestic production sales

12-fold, while exports will have to increase almost six times, according to PwC.

There also needs to be improvement in relations between the industry

and government, particularly with research-based companies, most of them

multinational players. Companies have been critical of the power wielded over

the sector by the Turkish Ministry of Health, which accounts for more than

three quarters of drug purchases.

Earlier this year, AIFD accused the government of not doing enough to

encourage investment and being too focused on cutting drug prices. Now, as

the government works on an action plan for the sector, ties between the two

sides have become closer. Kadir Tepebasi, AIFD vice chairman, said in July at

the BIO International Convention in Boston, “We are very happy and excited”

that the government and the research-driven segment had the same long-term

targets and approaches.

Nihat Ergun, the minister of science, industry, and technology whose

department is responsible for the development of the pharmaceuticals sector,

has revealed that the strategy document due to be finalized by the end of this

year will highlight R&D assistance, investment in human resources, and more

backing for drugs exports.

The PwC and BCG reports propose that the industry can reach ambitious

targets by 2023—if the appropriate steps are taken. A substantial increase

in domestic and foreign investment in the sector is required, especially in

production capacity for high value-added medicines, according to PwC. BCG

stresses the importance of the development of production and R&D clusters.

Over the next 10 years, the objective of more than doubling pharmaceutical

production to $23 billion, raising exports from approximately $520 million

last year to approximately $7 billion, turning a current trade deficit of more

than 80% into a net surplus, and increasing R&D investment from around

$100 million to $1.7 million “is not just a dream,” says Guldem Berkham, AIFD

president.

Both the industry and the government believe that the country has several

advantages that provide a platform for the rapid growth of the pharmaceutical

sector. The country’s domestic-drugs market has more than doubled over the

past 10 years, while with a population of 75 million, its per capita medicines

consumption is still relatively low. Over the past decade, visits to primary care

Discovery Pipeline

November 2012 www.biopharminternational.com BioPharm International 7

Global News

Page 10: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

facilities have more than tripled and to hospitals more than

doubled, according to BCG.

Turkey has a strong infrastructure for medicines

manufacture with around 300 pharmaceutical producers,

making it the seventh largest pharmaceutical industry in

Europe and 16th in the world. The country has well-established

universities providing a basis for R&D activities.

Above all, Turkey is strategically located at the crossroads

between Europe and Asia. It has the logistical connections to

supply Europe, which has annual pharmaceutical imports of

$264 billion; Russia with imports of $11 billion; Central Asia and

the Caucasus with $2 billion; and the Middle East and North

Africa with $14 billion.

To realize the potential of these benefits by attracting

large amounts of investment, the industry argues that the

government must first introduce a more sustainable drugs

pricing policy. Currently, Turkey’s medicines prices are among

the lowest in the world.

The country has had some recent investment successes.

These include Amgen’s $700-million takeover of Mustafa

Nevzat Pharmaceuticals, a Turkish generics producer; the

formation of a partnership between Otsuka of Japan and

Abdi Ibrahim of Turkey; and a decision by Zentiva, a Czech

subsidiary of Sanofi, to make Turkey a production base for

generic drugs.

A survey earlier this year by AIFD, however, found that price

cuts by the government have been holding back investment.

More than half of 23 research-based pharmaceutical

companies said they had had to cancel investments because of

government measures, mainly on prices.

Another problem has been a decision by the government

not to recognise GMP certificates from the EU and the US.

Instead, foreign production facilities have to be inspected

first by Turkish officials before Turkish marketing licences for

medicines can be granted.

An AIFD survey in April 2012 found that approvals of almost

300 medicines of foreign research-based companies were

being delayed because of the lengthy time it was taking to

gain GMP certification. “The GMP issue has to be sorted out

through mutual recognition agreements with the EU and US,”

says an IEIS official.

Another drawback for Turkey is a poor reputation for IP

protection in which it is rated 70th in the world. In some key

areas, the country has a lot of ground to make up before

its pharmaceutical manufacturing industry can attain a top

global ranking.

References  

1. PwC and AIFD, Turkey’s Pharmaceutical Sector—Vision 2023

Report, Strategy Document (Sept. 2012).    

2. Boston Consulting Group, Partnering with the Government

to Globalize the Turkish Pharmaceutical Industry (Nov. 2011).

—Sean Milmo is a freelance writer based in Essex, UK

On Oct. 22, 2012, a consortium in France announced the

establishment of Europe’s first ever industrial manufacturing

facility dedicated to the large-scale production of novel,

advanced cell-based therapy medicinal products. The

plant, located in Les Ulis, near Paris, at the site of the French

biopharmaceutical company LFB Biotechnologies, has the

capacity to produce 3000 to 5000 therapeutic batches per

year. The facility will be used for innovative R&D projects on

autologous and allogeneic cell therapies that are progressing

from early clinical research stage up to industrial production.

The facility project, known as C4C, is coordinated by

CELLforCURE, a subsidiary of LFB Biotechnologies, and will

involve two biotech companies—Celogos and CleanCells—as

well as seven public organizations and university medical

centers. Members of the consortium have invested US$104

million in the project alongside US$39 million public-

sector financial aid provided by OSEO, France’s state

innovation agency. The project seeks to provide an efficient

pharmaceutical and industrial tool for improving patient

access to cell therapies. Academic, public and private sector

stakeholders will have access to the new facility for routine

production of phase III clinical trial batches and commercial

batches of new cell-therapy products.

Five products are currently under development, with the

first batches expected in late 2013, according to a press release:

t� Bordeaux University Medical Center is collaborating

with France’s National Blood Service (EFS) on the GRAPA

program.

t� The CEL-02 cell therapeutic, developed by Celogos, is

undergoing Phase II evaluation.

t� Autologous islet of Langerhans grafts for the treatment

of post-pancreatectomy diabetes is in Phase I/II

development.

t� A cell-based immunotherapeutic, developed at the Cell

and Gene Therapy Unit at Nantes University Medical

Center, is undergoing Phase III evaluation.

t� The Mesenchymal and Myocardial Ischemia program,

conducted by Toulouse University Medical Center in

partnership with the EFS to evaluate the potential use of

mesencymal stem cells from bone marrow in combating

left ventricular myocardial ischemia, is in Phase II

development.

The industrial manufacturing facility will be validated

through the production of these five innovative cell

therapeutics and aims to meet the criteria set out by the

European and North American health authorities and

regulatory agencies. In addition to the C4C project’s

own cell-therapy products, the facility will offer custom

manufacturing services.

—Adeline Siew

Europe Establishes First Facility for Cell-Based Therapeutics

8 BioPharm International www.biopharminternational.com November 2012

Global News

Page 11: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

Biomanufacturing “out of the box“FlexMoSysTM – flexible, modular and fully integrated solutions

Page 12: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

10 BioPharm International www.biopharminternational.com November 2012

Regulatory Beat

Dig

ita

l V

isio

n/G

ett

y I

ma

ge

s

FDA officials are mapping out procedures

and policies for establishing an abbrevi-

ated pathway for testing and developing

safe and effective biosimilar products. Because

the regulatory framework for conventional

generic drugs does not fit these more complex

products, FDA is adopting a “weight of evi-

dence” approach for evaluating biosimilars,

starting with extensive comparative analysis

and functional studies to determine what pre-

clinical and clinical studies may be required.

The “residual uncertainty” that remains from

physical analysis is key to determining the

size and scope of further testing, explained

Janet Woodcock, director of the Center for

Drug Evaluation and Research (CDER), at a

September conference on biosimilars spon-

sored by FDA and the Drug Information

Association (DIA). Woodcock noted that this

heavy reliance on physical comparison rep-

resents “a paradigm shift” in how FDA docu-

ments drug safety and efficacy.

FDA la id out it s basic approach for

biosimilar development in three draft guid-

ance documents publ ished in February

2012 , and a CDER/Center for

Biologics Evaluation and Research

(CBER) Biosimilar Implementation

Committee is weighing comments

and further changes. Drug-review

staff members are talking to manu-

facturers about biosimilar develop-

ment options, as seen in 47 requests

for pre-investigational new drug

(IND) meetings on proposed biosim-

ilars to 11 reference products, as of

early October. Agency staffers have

held 30 meetings, and 12 INDs have

been filed for possible biosimilars,

some involving new development

programs and some building on data

from products already licensed in Europe. This

advisory program is highly work-intensive

for FDA, because each meeting with a spon-

sor entails at least four internal sessions and

consultation with CDER and CBER biosimilar

review committees to ensure that staff advice

is consistent across divisions.

Amidst all the discussion, the agency still

awaits its first official 351(k) biosimilar appli-

cation, as authorized by the Biologics Price

Competition and Innovation Act (BPCI),

which was enacted through the Affordable

Care Act of 2010. Teva Pharmaceuticals

recently received FDA approval for its ver-

sion of Amgen’s Neupogen (filgrastim), but

opted for a biological license application (BLA)

instead of the new route. This approach gives

Teva 12-years exclusivity for its product and

avoids the complex patent dispute resolution

process established by BPCI.

Added resources to support FDA meetings

and advice will come from the Biosimilar

User Fee Act (BSUFA), which was included in

the July 2012 FDA Safety and Innovation Act

(FDASIA). It authorizes fees for biosimilars that

are similar to those for new drugs and biolog-

ics, but “frontloads” the program by allowing

FDA to collect a Biosimilar Biological Product

Development (BPD) fee—10% of the nearly $2

million application fee—when a sponsor holds

its first official meeting with FDA or files an

IND. Another 10% is paid every year that the

IND remains active, but total BPD fees will be

subtracted from a future BLA application fee

so that the final payment remains the same as

for other new drugs.

In addition to technical and scientif ic

challenges, a number of legal and regula-

tory issues will determine the development

path for a new biosimilar. A convoluted pro-

cess for addressing exclusivity and patent

Product Analysis Key to Biosimilar DevelopmentManufacturing and in-depth characterization provide basis for demonstrating product equivalence.

Jill Wechsler is BioPharm

International’s Washington editor,

Chevy Chase, MD, 301.656.4634,

[email protected].

Read Jill’s blogs at

PharmTech.com/wechsler.

Page 13: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 11

Regulatory Beat

Hot-Topic Roundup

Crackdown on online pharmacies

FDA is urging consumers to recognize the dangers of online drug

purchases through a new “BeSafeRx” campaign. At the September

2012 meeting of the Partnership for Safe Medicines, Commissioner

Margaret Hamburg cited data on the rising purchase of drugs through the

Internet and FDA’s efforts to help the public identify fraudulent operators.

Hamburg and others also highlighted the latest attack on illegal web-

based pharmacies, which was coordinated by Interpol last month. The

fifth “Operation Pangea” resulted in the shutdown of more than 18,000

pharmacy websites worldwide and seizure of approximately $10 million

worth of drugs. In the US, FDA took action against almost 4000 online

pharmacies, sending out Warning Letters, seizing illegal products, shut-

ting websites, and seeking civil and criminal penalties.

Meanwhile, talks continue on Capitol Hill about policies for establish-

ing a system for tracking drug shipments through the supply chain. FDA

and industry still hope to devise a workable national track-and-trace

system, but have been stuck over whether to start with a more limited

batch-level tracking approach versus the feasibility of requiring tracking

to vial level, as FDA prefers.

Promoting antibiotics

FDA announced the formation of the Antibacterial Drug Development

Task Force on Sept. 24, 2012, as required by provisions in the July 2012

FDA Safety and Innovation Act (FDASIA) that encourages research and

development of new antibiotics. This group of Center for Drug Evaluation

and Research (CDER) scientists and clinicians will develop additional

guidance, explore new research methods, and define factors that have

limited the pipeline for therapies in this area. FDASIA also offers extend-

ed exclusivity for new antibacterial and antifungal drugs that meet certain

requirements, offering hope that new drugs to combat infectious disease

will also limit resistance to existing treatments, which has become a seri-

ous global issue.

More focus on drug shortages

Among other efforts to combat drug shortages, FDA is elevating the sta-

tus of a special Drug Shortages Staff (DSS) in CDER. DSS will move from

CDER’s Office of New Drugs to report directly to CDER Deputy Director

Doug Throckmorton, with an eye to better coordinating a number of new

provisions in FDASIA that require expanded manufacturer notification of

situations that could lead to shortages.

The drug shortage crisis appears to be abating, as a number of

troubled facilities producing generic sterile injectables have recovered,

and new ones have come online. DSS Associate Director Valerie

Jensen recently reported, in a presentation at the September meeting

in Washington of the American Osteopathic Association, that FDA

had tracked only 100 shortages as of late September, compared to

more than 180 during the same period last year, when shortages

totaled 251. Moreover, FDA says it prevented 195 shortages in 2011

and almost 100 in the first half of this year. More high-quality manu-

facturing gets credit for the improvement, along with FDA efforts to

encourage alternative supplies.

Further shortage prevention strategies may come from DSS analysis

of links between shortages and inspections or regulatory actions, and

whether there were signals of problems (e.g., field alerts, product recalls,

similar events overseas) that could have alerted FDA earlier to a looming

supply problem.

Congress tackles cargo theft

In September, Congress quietly enacted legislation that increases

penalties for crimes involving theft and hijacking of drugs, biolog-

ics, and medical devices. The House approved the Safe Doses

Act (HR 4223) in June. The Senate approved the Act in late

September, and President Obama signed the bill into law last

month. Manufacturers have long complained that criminals face

stiffer penalties for stealing handbags and CDs than prescription

drugs, and the legislation aims to change that by imposing stiff

fines and jail sentences on offenders. The measure, which was

introduced by Rep. James Sensenbrenner (R-WI) in March, and

sponsored by Sen. Charles Schumer (D-NY) in the Senate, impos-

es penalties for falsification, alteration, and forgery of medical

products and their subsequent transportation or sale.

Avoiding budget cuts?

The prospect of across-the-board federal budget “sequestration” to

reduce the projected $1.2 trillion budget deficit is prompting a host

of savings proposals from interest groups and policy analysts. The

Pharmaceutical Care Management Association (PCMA), which rep-

resents pharmacy benefit managers (PBMs), weighed in last month

with a list to cut federal spending on prescription drugs by $100

billion over 10 years. PCMA’s main strategies are to promote more

generic drug use by Medicaid and Medicare Part D drug plans, to

encourage mail order services for elderly patients at home, to reduce

Part D “protected drug classes” that limit price negotiations for cer-

tain drugs, and to end the tax deduction for direct-to-consumer drug

advertising.

Recently issued guidance documents

t� *OJUJBM�$PNQMFUFOFTT�"TTFTTNFOUT�GPS�5ZQF�**�"1*�%SVH�.BTUFS�'JMFT�

VOEFS�(%6'"�'%" �%SBGU �0DU��� ������

t� 4UBCJMJUZ�5FTUJOH�PG�%SVH�4VCTUBODFT�BOE�1SPEVDUT�GPS�"/%"T�'%" �

%SBGU�4FQU���� ������

t� 4FMG�*EFOUJGJDBUJPO�PG�(FOFSJD�%SVH�'BDJMJUJFT �4JUFT�BOE�0SHBOJ[BUJPOT��

'%" �%SBGU �"VH���� ������

t� (FOFSJD�%SVH�6TFS�'FF�"NFOENFOUT �2VFTUJPOT���"OTXFST�'%" �

%SBGU �"VH���� ������

t� %FWFMPQJOH�%SVHT�GPS�5SFBUNFOU�PG�"DVUF�#BDUFSJBM�4JOVTJUJT�'%" �'JOBM �

0DU���� ������

t� %FWFMPQJOH�%SVHT�GPS�5SFBUNFOUT�PG�"DVUF�#BDUFSJBM�0UJUJT�.FEJB�

'%" �'JOBM �0DU�������

Page 14: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

12 BioPharm International www.biopharminternational.com November 2012

Regulatory BeatRegulatory Beat

rights presents a clear challenge,

and there is considerable debate

over biosimilar names and prod-

uct codes. The size and shape of

clinical studies remains uncer-

tain, along with questions about

non-US comparators and bridging

studies. Interchangeability is the

ultimate goal for most biosimi-

lars manufacturers, but FDA indi-

cates that such a designation may

require more extensive test data.

All sides anticipate lawsuits, citi-

zens’ petitions, and other legal

battles to resolve these and other

controversies.

ANALYTICS FIRST Despite these uncertainties, the

first step for manufacturers is to

develop a clear analytical plan for

demonstrating biosimilarity of

a follow-on to a reference prod-

uct. A “step-wise” approach for

doing this was laid out by CDER

officials at the DIA meeting and

subsequently by Steve Kozlowski,

d i rec tor of CDER’s Of f ice of

Biotechnology Products (OBP),

at the Generic Pharmaceutical

Association’s (GPhA) fall 2012

conference. Kozlowski observed

that manufacturing and product

analysis are usually at the “low

end of the totem pole” in drug

development, but that a “total-

ity of evidence,” starting with

structural and functional char-

acterization, provides a founda-

tion for determining the scope

of preclinical and clinical studies

for a biosimilar. “Once you know

how close you are,” Kozlowski

explained, “the rest follows.”

To make the most of an ini-

tial advisory meeting with FDA,

sponsors should have a clear

rationale for product develop-

ment, with early characterization

data for the proposed biosimilar

and reference product lots and

justification for as much of the

analytical approach as possible.

In-depth characterization assay

development is desirable, as are

preliminary analytical functional

similarity studies and formulation

studies. It also may help to have

validated research and stability

assays to support an IND.

“Know your protein” is a main

FDA theme for biosimilar develop-

ment. Selection of analytical test

methods should be based on the

nature of the protein and knowl-

edge of its structure. Agency staff

wants to know which attributes

are important and how the rela-

tionship between protein attri-

butes and the clinical safety and

efficacy profile can predict “clini-

cal similarity.” Also important

is whether differences between

a chosen expression system and

that of the reference product will

significantly affect process- and

product-related substances and

impurities, how differences in

the impurity profile or in excip-

ients will affect safety, and the

strengths and weaknesses of each

analytical method.

FDA’s draft guidance on qual-

ity considerations for biosimilars

describes a broad range of analyti-

cal studies that may be relevant,

including assessment of expres-

sion system, physicochemical

properties, functional activities,

receptor binding, immunochemi-

cal properties, impurities, ref-

erence product, and stabil ity.

Protein evaluation stands to ben-

efit from a growing number of

analytical tools, including mass

spectrometry, peptide mapping

and chromatography to assess

amino acid sequence, protein fold-

ing, subunit interactions, hetero-

geneity, glycosylation, PEGylation,

bioactivity, and other methods.

FUTURE FINGERPRINTING?Advances in manufacturing sci-

ence and adoption of quality-by-

design approaches may support

comparative assessment using a

fingerprint-like analysis or “super

characterization” approach that

involves evaluating combinations

of attr ibutes using orthogonal

methods. It is not clear whether

biotech manufacturing processes

are too variable to allow for a fin-

gerprinting approach, Kozlowski

explained at the GPhA confer-

ence, but he ant ic ipates that

upfront efforts to select appropri-

ate cell lines will permit man-

ufacturers to deliver an exact

desired product.

Transparency also is important.

It is better for a manufacturer to

acknowledge uncertainties from

test results and to seek advice

from FDA for dealing with these

issues than to ignore or hide

d i s c r e p a nc ie s . F DA adv i s e s

manufacturers to carefully con-

sider the importance of differ-

ences in expression systems

and the need to justify minor

modifications in an amino acid

sequence.

At the same time, FDA is trying

to be flexible, Kozlowski empha-

sized, noting that differences in

formulation from the reference

product may be acceptable, as

well as alternative delivery devices

or container-closure systems.

Applicants may market a proposed

biosimilar for fewer than all con-

ditions of use and presentations

for which the reference product is

licensed.

“The more analytical data one

has up front, the more targeted

the rest of development can be,”

according to Koslowsky. Although

FDA expects that sponsors will

have to conduct at least one

immunogenicity study, Koslowski

says that “the door is open,” to

the concept that “added studies

may not be necessary.” ◆

Page 15: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

Parenteral Drug AssociationTraining and Research Institute (PDA TRI)

Customized Training at Your Facility or Ours

PDA TRI can work with you to bring custom “in-house” training solutions

and expert instructors right to your doorstep. Get training solutions that

meet your specifi c needs while saving time and money on travel. If your

offi ces are not conducive to training, PDA TRI can host your programs in

our facility located in Bethesda, MD. Our state-of-the-art facility includes

an aseptic processing suite with a fi lling room, component prep lab

and gowning/degowning areas, clean-in-place lab, microbiology lab,

biotechnology lab, classrooms and student break areas.

We can deliver one of our existing training courses to your organization

or we can work with you to develop the custom training you need. Below

fi nd a list of available subject areas covered by TRI training.

Aseptic

Biotech

Environmental Monitoring

Filtration

Microbiology

Quality and Regulatory Affairs

Specialized

Training

Validation

We look forward to bringing our expert-led pharmaceutical

and biopharmaceutical training to you.

Contact us to discuss your specifi c needs:

Robert Dana

Senior Vice President Quality and Regulatory Affairs

and PDA Training and Research Institute

+1 (301) 656-5900 ext. 224

[email protected]

For more information please visit www.pda.org/courses

Page 16: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

14 BioPharm International www.biopharminternational.com November 2012

Perspectives on Outsourcing

Do

n F

arr

all/G

ett

y I

ma

ge

s

A revolut ion star ted about a dozen

years ago in the federal government

and is spreading to state, local, and

commercial purchasing organizations. This

revolution is about turning procurement

efforts into a sustainable cash-flow-generat-

ing stimulus machine by extending available

resources to get more spend under manage-

ment, competitively bid spend more often,

and take advantage of market dynamics that

create marginal pricing opportunities. It is

a change in thinking that drives maximum

efficiency, maximum savings, and creates a

truly holistic procurement management sys-

tem that goes well beyond the 50% to 80%

control that most organizations have.

Many leading companies and organiza-

tions have worked hard during the past sev-

eral years to become lean, agile, and flexible.

Among other initiatives, they have taken

a close look at support function staff ing

(including procurement) and have down-

sized, right-sized, outsourced, and off-shored

with the net effect resulting in an environ-

ment where employees are having to do more

with less resources or more without more

resources. Another set of com-

panies and organizations have

taken a different path, and, as it

relates to purchasing goods and

services, have never upgraded

the effort nor staffed their pro-

curement organizations appro-

priately. In either case, the net

outcome has been one where

real and significant savings have

been left on the table because

there were not enough resources

or not enough skilled resources

available to competitively bid a

maximum amount of the com-

pany’s or organization’s spend for goods and

services.

With the remaining resources, highly

skilled organizations take a position of focus-

ing on ensuring that crucial goods and ser-

vices are protected by contracts and rebid

every one to five years. These companies

leave the smaller spends to sort themselves

out with the help of dozens or even hun-

dreds of business end-users following pro-

curement policy to engage suppliers and

hope for the best. The companies that have

less skilled purchasing departments continue

to focus on transactional buying with little

professional oversight. In both cases, orga-

nizations are paying more than they need to

pay and also are putting their companies at

risk by not formally vetting many of the sup-

pliers they are using.

IMPROVING THE WHOLE In the effort to cut costs, many companies

in the pharmaceutical industry and other

industries have purposely taken an approach

that reduces cost by cutting infrastructure,

Organizations are paying more

than they need to pay and are

putting their companies at

risk by not formally vetting

many of the suppliers

they are using.

Creating a Holistic Procurement System The procurement organization rethinks sourcing for maximum efficiency and results.

Gregg Brandyberry is senior vice-

president of FedBid, and formerly

senior advisor for A.T. Kearney

Procurement and Analytic Solutions,

[email protected].

Page 17: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 15

Perspectives on Outsourcing

inc lud ing s ig n i f icant reduc-

t ions in ful l-t ime employees.

Although much of this cutting

is needed and laudable, there

are many examples where cuts

have been equally applied to all

internal functions (e.g., 15% for

all support staff). Although this

approach fixes one part of the

business (i.e., procurement labor

costs), it has a negative impact

on the organization as a whole.

Procurement is one of the few

functions that can have a direct

and ongoing impact on the bot-

tom line and should be staffed

or supported appropriately for

maximum savings delivery and

overall risk mitigation.

RETURN ON INVESTMENT THROUGH PROCUREMENT To offer an example of the value

of procurement, take the case of

a small but growing pharmaceu-

tical company that had no real

center-led procurement strategy

or organization. When it came

to purchasing indirect goods

and services (i.e., all goods and

services with the exception of

direct materials to make prod-

ucts), senior management and

administrative assistants became

the purchasing experts, which

resulted in little, if any, competi-

tive processes being used, suppli-

ers not properly being screened,

and generally creating unneces-

sary cost and risk for the orga-

n i zat ion. Poor proc u rement

practice was evident when buy-

ing contract research organiza-

tion services; marketing services;

facility services; maintenance,

repair, and operat ions; of f ice

supplies; benefits; and other ser-

vices.

A f te r r ev iew i ng t he com-

pany’s buying pract ices, l im-

ited policies, and several recent

purchases, it was clear that the

company was over-pay ing on

average by a minimum of 20%

for indirect goods and services.

This organization spent approxi-

mately $100 mil l ion per year

on indirect goods and services

with revenues of approximately

$250 mi l l ion. It was recom-

mended that the company make

an investment in upgrading its

procurement capability by nam-

ing a head of procurement to

institute a center-led program for

purchasing direct materials and

indirect goods and services. The

investment would easily have a

50 times return during an 18- to

24-month period, according to

conservative estimates.

EXTENDING THE REACH OF PROCUREMENTApproximately 10 years ago, fed-

eral agencies began using FedBid

to buy common goods and basic

services. FedBid is also available

for commercial use and offers

a highly efficient procurement

solution for commodity prod-

ucts and basic services. There is

never any cost to the participat-

ing sellers and no upfront cost

to the buying organizations. A

small transactional fee is added

to the winning sellers’ bid and

is paid by the buyer as part of

the purchased price. After the

buyer pays the sel ler, FedBid

col lects the transact ional fee

f rom the sel ler. Both par t ies

win; the sel ler receives addi-

tonal revenue and the buyer

nets average savings of > 10%

(based on more than a decade

of buying awards). If the prod-

uct or service is not currently

in the marketplace, the mar-

ketplace operator wi l l at, no

charge, find the right sellers and

even train a company’s existing

suppliers so they can compete

on a regular basis in the market-

place.

Today, the marketplace has

more than 57,000 approved sell-

ers. Buyers use a simple and

ef f ic ient Internet-based user

interface to enter the require-

ments of what they want to

buy (i.e., a specif ication), and

FedBid, being a fully-managed

marketplace, takes f u l l con-

trol of the sourcing process to

ensure robust competition from

multiple sellers, validate qual-

ity assurance of leading bids,

and delivers f inal sel ler pr ic-

ing options back to the buyer

for award. The marketplace is

fair, ethical, and transparent. It

enables dynamic competition by

providing the best market pric-

ing avai lable at the t ime the

purchase is completed. FedBid is

rapidly branching out into state

and local governments, schools

and universities, and commercial

organizations.

LOOKING AHEADEvery organization has an oppor-

tunity to do a better job of buy-

ing indirect goods and services.

Buying smart, sav ing money,

and reducing risk by having bet-

ter controls in place makes real

economic sense, especially in

today’s environment. z

Procurement is one of

the few functions

that can have a

direct and ongoing

impact on the

bottom line.

Page 18: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

16 BioPharm International www.biopharminternational.com November 2012

Compliance Notes

Ph

oto

dis

c/G

ett

y I

ma

ge

s

Supplier quality management (SQM) is,

at its core, a compliance risk-mitigation

strategy within the overarching strategy

of quality risk management (QRM). I think

sometimes, however, the emphasis becomes

too focused on the compliance risk asso-

ciated with the execution of the supplier

quality-management program itself and we

need to remind ourselves that one of the

most fundamental compliance risks SQM is

designed to mitigate is product failure. The

effectiveness of SQM in mitigating the risk of

product failure, however, is directly related

to how well you understand the impact the

material supplied has on product quality.

Absent that, one could have an SQM program

that is fully compliant in its execution and

yet ineffective in preventing product failure.

Application of quality by design (QbD) in

product development and/or postdevelop-

ment product/process characterization com-

bined with effective knowledge management

have proven to be a highly cost-effective

approach to rigorously connect raw mate-

rial characteristics to product critical quality

attributes. Integrating these tools and tactics

into your supplier quality-management sys-

tem can appear a daunting task, but

the payoff is well worth the effort.

Not only will you be able to focus

supplier management on the right

issues, you won’t squander your

resources and those of your suppli-

ers focusing on things unimportant

to product quality.

At one point in my career, I

accepted a manager ia l rotat ion

assignment in which I moved from

manager of pharmaceutical manu-

facturing to lead the procurement

group. As part of a review of overall sourcing

strategy, my new team identified a supplier

from whom we purchased a basic excipient as

a potential target for negotiation or replace-

ment because their cost seemed out of line.

Dialog with the supplier revealed that the

cost differential was being driven by our

specification that the product be packaged

in paper bags of an unusual size. Ironically,

from my previous job, I knew that handling,

opening, and loading those paper bags into

granulators was not only time consuming,

but we also ended up putting partial bags

back into the warehouse because their weight

wasn’t an even multiple of our batch size.

Having the supplier switch to providing the

material in totes eliminated the purchase-

cost differential and reduced our conversion

cost. We had essentially been paying a sup-

plier extra money to provide us with some-

thing we did not need.

So, what does this have to do with QbD,

product knowledge, and supplier quality

Having a supplier maintain

tight control over a

characteristic that is not

really important to the

quality of your product

often drives extra cost.

J. Paul Catania is a managing

consultant at Tunnell

Consulting, jpaul.catania@

tunnellconsulting.com.

Connecting QbD, Knowledge Management, and Supplier Quality ManagementUnderstanding overall supplier capability versus the critical-to-quality attributes of your product can reduce both risk and cost.

Page 19: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological
Page 20: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

18 BioPharm International www.biopharminternational.com November 2012

Compliance Notes

management? Hav ing a sup -

plier maintain tight control over

a characteristic that isn’t really

important to the quality of your

product often drives extra cost

which they, naturally, pass on

to you. Incoming quality con-

trol and supplier quality man-

agement efforts to ensure they

meet the unnecessary specifica-

tion add to the overall conver-

sion cost. Worse, when achieving

the specification is difficult for

the supplier, compliance r isk

increases because, unnecessary

or not, failure to meet the speci-

fication is a deviation.

Meanwhile, in the absence of

process characterization rigor-

ously linking raw-material char-

acter ist ics to product quality,

you risk under-specifying some-

thing whose negative impact you

would gladly pay your supplier

extra to avoid, and on which

focusing supplier quality-man-

agement resources makes good

business and compliance sense.

Consider the basic supplier

qua l it y-management consid -

eration of multiple versus sole

sourcing. A colleague recently

reminded me of Deming’s pref-

erence for sole sourcing. After

all, the resultant variability of

multiple suppliers can’t be any

less than the most va r iable

among them, and the probabil-

ity is that the composite vari-

ability will be greater than any

of them alone. Meanwhile, the

cost to manage each of them is

incremental. When the risk of

supply interruption is unaccept-

ably high, however, the incre-

menta l suppl ier-management

cost of qualifying another sup-

plier is justified. Ironically, even

when the secondary supplier is

duly audited and qualified, when

finally utilized, supply continu-

ity is often jeopardized by unex-

pected dev iat ions in product

quality.

Even though the secondary

supplier’s product meets all the

specifications, the centering or

variability of some characteris-

tic to which the process is sen-

sitive isn’t the same as that of

the primary supplier and, there-

fore, isn’t the same as what was

used to develop and validate the

process. Sometimes, it’s a char-

acteristic never previously identi-

fied as important and for which

you currently don’t even have a

specification. Far more often, it’s

a specified characteristic whose

variability has more impact on

product quality than previously

understood.

The risk of this quality devia-

t ion happening is h igher in

products developed using tra-

ditional three-batch validation.

However, it can occur even in

products developed using QbD,

par t icu la rly when developed

using raw material f rom only

one supplier because suppliers

are only shipping product within

specifications and attempting to

minimize variability. Consider a

supplier who your supplier qual-

ity-management team has quali-

fied against specifications. The

supplier’s natural process capa-

bility may result in the value of

a critical-to-quality characteris-

tic being centered to one side of

the specif ication but within a

narrow enough range that what

it produced generally meets the

specification. When it doesn’t, it

sells it to an alternate industry.

Often, suppliers are unwilling

or even unable to produce mate-

rial much outside their normal

process capability. So, the prod-

uct knowledge initially devel-

oped using a g iven supplier ’s

material will often be narrower

than what you need to push the

limits of your design space. Over

time, however, there will prob-

ably be instances in which you

will receive lots inside your spec-

ification but outside your orig-

inal design space, particularly

as you seek to qualify second-

ary suppliers. Effective knowl-

edge management will allow you

to identify and integrate these

lots into your design space and

expand it.

The key message here is that

for these risk-management strat-

egies to effectively inform sup-

p l i e r qu a l i t y m a n a ge me nt ,

product and process understand-

ing can’t be viewed as a once-

and-done static event completed

during initial product develop-

ment. Rather it must dynami-

cally evolve and grow over the

entire course of a product’s life-

cycle. This growth is part icu-

larly important when it comes

to supplier quality management

because your supplier’s processes,

like yours, are subject to process

variability and drift. Gathering

and analyzing data on an ad hoc

For risk-management strategies to

effectively inform supplier quality

management, product and process

understanding can’t be viewed

as a once-and-done static event.

Page 21: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 19

Compliance Notes

basis to characterize a process

in reaction to product failures

is sometimes unavoidable. More

and more, however, the bench-

mark pract ice is to recognize

and support the total lifecycle

evolution of product and process

knowledge through proact ive

development of knowledge-man-

agement systems designed to

dynamically integrate raw mate-

rial and product release testing

data with data within manufac-

turing batch records.

The good news is that whether

or not your product was devel-

oped using QbD, where it is in

its l i fecycle, or how sophist i-

cated your current knowledge-

management system, it’s never

too late to characterize processes

and develop product and process

knowledge you can leverage to

mitigate risk and focus resources

to maximize cost effectiveness

in supplier quality management

and in QRM overall. We have

seen organizations quickly and

significantly reduce risk and cost

through process character iza-

tion in reaction to product fail-

ure. Proactive characterization

and knowledge management is

just as effective at reducing risk

and costs far less because it isn’t

initiated by a product deviation

crisis. Moreover, a knowledge-

management system need not be

a complex and expensive infor-

mat ion systems under tak ing.

When the true value of integrat-

ing data already being generated

is demonstrated through these ad

hoc characterizations, organiza-

tions often identify simple ways

to leverage what they learned on

an on-going basis.

I f you don’t want to wa it

for product failure to catalyze

action, an overall portfolio risk

assessment will inform your pri-

orities vis-à-vis which products

you target first for characteriza-

tion. This can be as simple as a

review of product, process, and

test ing deviat ions to identify

the products which give you the

most trouble. Given the connec-

tion between SQM and product

knowledge, a holistic approach

that includes an understanding

of overall supplier capability ver-

sus critical-to-quality attributes

is the way to go. Along the way,

your organization will not only

gain product knowledge that

can be leveraged to reduce both

risk and cost, you will inevita-

bly discover opportunit ies to

connect and leverage informa-

t ion a lready being generated

and begin to build the founda-

tion of your knowledge-manage-

ment system. ◆

4701 Southport Supply Rd., SE, Suite 7

Southport, NC 28461

Phone: 910-454-9442��%���ax: 910-454-9443

Trust The Experts

Email: [email protected]

A full menu of kits

available for…

Microbial HCPs

�� E.coli

�� ����������� �

�� ��������������������

�� ������������������

�� � � �������� �

�� Sac������������� � ���

Transgenic Applications

0� Bovine Plasma Proteins

0� Goat Casein

0� Goat IgG

0� Total Goat Milk Proteins

Mammalian

Cell Line HCPs

0� CHO 0�NS/0

0� A549 0�PER.C6®

0� BHK 0�SP2/0

0� HEK 293 0�Vero cell line

0� MRC5

/� �����/���������/�������C

/� Western Blot Kits

/� A0+&.���&1�#��

�+&!,#&��

/� C,+'% �#��+&!,#&���

,��)( )&+����,���, ��,

�&,&+�������

/�Custom Development

���vices for process

���"&1"� + ).��

F,������

y,���,���ell

contaminant

+��#�:

Page 22: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

20 BioPharm International www.biopharminternational.com November 2012

The Disposables Advisor

Alth

ea T

ech

no

log

ies

Jerold Martin is the senior vice-

president of global scientific

affairs at Pall Life Sciences,

Port Washington, NY, US.

516 801 9086, jerold_martin@pall.

com. Jerold is also the chairman

of the board and technology

committee of the Bio-Process

Systems Alliance.

Tubing is a crucial part of any single-

use system and is required for secure

fluid transfer. However, it also presents

a significant contact area that can poten-

tially contribute to adsorption, leachables,

or particulates, so selection of appropriate

tubing for each application is important.

The best starting point is to consider the

process f luid and conditions to which the

fluid transfer tubing will be exposed. In this

column, I will also consider the potential

impact on product, the types of tubing avail-

able to choose from, and other consider-

ations in making a selection.

The recommendations below are based

on my experience with single-use systems,

along with suggestions by technical experts

from three tubing suppliers: Steve Wilkowski

of Dow Corning, John Stover of NewAge

Industries, and Christopher Shields of Saint-

Gobain.

IMPACT OF FLUID AND PROCESS CONDITIONSThe first factor to consider when

selecting tubing is the nature of the

fluid you want to transfer. In sin-

gle-use bioprocessing, fluids range

from proteins or other biological

solutions to strong pH adjusters and

buffers. Biologicals may be sensitive

to ultraviolet light or oxygen, and

require opaque or low gas perme-

ability tubing. Acids and bases must

be used with chemically compatible

tubing. In all cases, it is necessary

to select tubing grades with low

extractables to ensure minimum

levels of leachables.

Once biolog ica l stabi l it y and

chemical compat ibi l ity require-

ments are defined, processing con-

dit ions should be considered, including

process temperature, pressure, t ime, and

sterilization conditions prior to use. Single-

use bioprocesses are generally conducted at

temperatures below 40  °C, so most tubing

options are compatible. If tube welding and/

or sealing is required on the system then

meltable tubing will be needed, whereas

the use of only quick-connects or ster-

ile connectors eliminates this temperature

requirement. Similarly, if the tubing is to be

autoclaved, it should be resistant to at least

125 °C.

The next process condition to consider is

pressure. Although the low-pressure limits of

single-use polymer-film biocontainers gener-

ally limit pressure requirements for system

tubing, tubing used upstream of sterilizing

filters that are integrity tested prior to use is

an exception. Depending on the grade of fil-

ter and integrity test method, test pressures

can range from 40  psi (2760  mbar) to 70  psi

(4825 mbar), or even higher with some myco-

plasma or virus filters. To accommodate this,

high durometer, braided or otherwise rein-

forced tubing must be incorporated upstream

of the filters to ensure suitable pressure rat-

It’s an Election Year.... Are You Making the Best Choices in Single-Use TubingJerold Martin considers the types of tubing available to the industry and how to make an informed selection.

Tubing used upstream of

sterilizing filters, and in final

filling, should be considered

for low silicone oil, along with

other leachables.

Jerold Martin

Page 23: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 21

The Disposables Advisor

ing for integrity testing. Another

exception is where peristalt ic

pumping is used to drive the

process f luid. Tubing selection

must consider the strength of

the tubing to provide consistent

flow and low generation of par-

ticulates. Tubing grades recom-

mended for peristaltic pumping

are typically qualified for much

longer serv ice than would be

applied in most single-use appli-

cations, but failure to consider

pumping suitability after ster-

ilization can lead to significant

problems.

Sterilization conditions must

be considered when select ing

tubing. Most single-use applica-

tions are gamma irradiated for

sterilization and consequently

require gamma stable tubing

(generally up to 50  kGy dosage),

but it is not uncommon to apply

autoclave sterilization for some

tubing mani folds and f i lt ra-

tion systems, especially during

process development and early

phase clinical batch manufac-

turing. The desired sterilization

method must be considered in

tubing selection. Information on

gamma sterilization is provided

in the BPSA Guide to Irradiation

and Sterilization (1).

IMPACT OF TUBING ON PROCESS FLUIDSThe pr imary concern for the

tubing’s impact on process f lu-

ids are biocompatibility, leach-

ables, adsorpt ion–absorpt ion,

and permeabi l ity of gas and

l ight (part icularly ultrav iolet

light). Biocompatibility is typi-

cally assessed by applying the

USP Biological Reactivity Tests

to the tubing material after a

“worst case” sterilization process,

either in vivo (<87>, cytotoxic-

ity) (2), or in vitro for Class VI

(<88>, implantable) plastics (3).

Other standard tests may include

pyrogenicity. Recommended stan-

dard reference tests for tubing are

described in the BPSA component

quality test matrices Guide (4).

Potential leachables are ini-

t ial ly assessed by considering

extractables determined under

exaggerated process conditions,

such as higher temperatures,

more aggressive solvents, and

longer contact times. The maxi-

mum sterilization process con-

ditions should also be included

(typically >125  °C steam auto-

clave or 50  kGy gamma irradia-

tion) because these factors can

increase the level of process

leachables f rom some tubing.

Similarly, the impact of heat

welding on leachables should

also be included in extractables

assessments. For more informa-

tion on determination of tub-

ing extractables data, see the

2008 and 2010 BPSA extractables

guides (5, 6).

Adsorption, where target mole-

cules bind to tubing contact sur-

faces, is a primary concern with

protein molecules, but the rela-

tive small-surface area to volume

ratio typically limits protein con-

centration losses to only highly

d i lute solut ions. Absorpt ion,

where target molecules migrate

into the tubing solid phase itself,

are a greater concern with small

molecules such as preservatives,

which are generally limited to

final formulations, but should be

... it is not uncommon

to apply autoclave

sterilization for some

tubing manifolds and

filtration systems.

DASGIP –

Parallel Bioreactor Systems

for Unparalleled Results.

As the industry leader for benchtop bioprocess-

ing solutions, we have defi ned the state of the art

in Parallel Bioreactor Systems. Our best-in-class

confi gurable control systems coupled to innova-

tive information management solutions support

interconnectivity to 3rd party offerings.

DASGIP –

We know bioprocessing – since 1991.

an EppendorfCompany

Page 24: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

22 BioPharm International www.biopharminternational.com November 2012

The Disposables Advisor

assessed for tubing used at that

stage of the process. Low per-

meability tubing (for gases and

light) can also provide reduced

absorption of small molecules.

A lthough par t ic les in tub -

ing used in upstream processes

(media, buffers and intermedi-

ates) upstream of filters have not

been a major concern, apply-

ing single-use systems to asep-

tic vaccine manufacturing or in

post-filtration formulation and

filling of biopharmaceuticals has

raised concerns about the poten-

tial impact of particles from tub-

ing fill lines on final dosages.

Extrusion of tubing is generally

a low particle-generating process,

but handling, cutting, joining,

and environmental conditions

for single-use system assembly

can have an impact. Particle lev-

els from tubing in systems used

downstream of f inal f iltration

should be considered in critical

applications.

TYPES OF TUBING FOR SELECTIONThe most common t y pes of

t u b i n g u s e d i n s i n g l e - u s e

appl icat ions are si l icone and

t h e r m o p l a s t i c e l a s t o m e r s .

Silicone tubing has a long his-

tory of safe use for a broad range

of bioprocess fluids, and provides

high temperature and gamma

radiation stability. Other ben-

efits include f lexibility, trans-

lucency, biocompatibility, and

smooth bore for low adsorption

and particulates. Peroxide-cured

grades have been successfully

used in laboratory and clinical

applications, but platinum-cured

silicone is preferred for single-

use systems because of lower

extractables. Braided grades are

available for high pressure appli-

cations. Silicone tubing cannot

be thermo-welded and is limited

to pre-assembled systems or con-

nections using quick-connects or

sterile connectors.

For tube welding and sealing

applications, thermoplastic elas-

tomer (TPE) tubing is required.

These are proprietary formula-

t ions, commonly refer red to

under trade names, and have a

good record of safe use and are

a lso prefer red where reduced

adsorption, absorption or gas

permeability is desired, though

they do not have the transpar-

ency of silicone tubing. Other

specialty formulations of tubing

are also available.

Both si l icone and thermo-

plastic elastomeric tubing are

manufactured in numerous pro-

pr ietary formulat ions, result-

ing in d i f ferent extrac tables

prof i les and other propert ies.

A case study was reported sev-

eral years ago on the effect of

tubing material on the bubble

point va lues of downst ream

s t e r i l i z i n g - g r a d e f i l t e r s . ( 7 )

Poly(dimethlysiloxane) silicone

oil leached from some grades of

tubing (both silicone and ther-

moplast ic elastomer) and was

adsorbed on downstream f i l-

ter surfaces. This reduced the

critical surface wetting energy

of the f i lter membranes, thus

impacting post-use bubble-point

values, causing false test failures.

Tubing used upstream of steril-

izing filters, and in-final filling,

should be considered for low

si l icone oil, along with other

leachables.

It is common for new users of

single-use systems to consider

choosing whatever grade of tub-

ing they have experience with in

their laboratory or clinical-batch

processes, but it is also impor-

tant to consider the selection

and sourcing of tubing by the

single-use system integrator. It

is also advantageous for integra-

tors to limit their approved tub-

ing sources, including inventory

control, cost-efficiency, and gen-

eration of additional qualifica-

tion studies (e.g., post irradiation

extractables). Good integrators

will also establish quality agree-

ments for traceability and sourc-

ing of raw materials (e.g., resins

and elastomers), change notifica-

tion protocols, and conduct sup-

plier audits. Consequently, once

your f luid and process require-

ments are defined, it can be ben-

eficial to consider suitable grades

among those already selected

and qualified by the single-use

system integrator.

REFERENCES1. BPSA, Guide to Irradiation and

Sterilization of Single-use Systems

(2008).

2. USP 29–NF 24 General Chapter <87>,

“Biological Reactivity Tests, in vitro,”

2525.

3. USP 29–NF 24 General Chapter <88>,

“Biological Reactivity Tests, in vivo,”

2526.

4. BPSA, Component Quality Test Matrices

Guide (2007).

5. BPSA, Guides to Extractables and

Leachables from Single-use Systems

(2008).

6. BPSA, Guides to Extractables and

Leachables from Single-use Systems

(2010).

7. B.K. Meyer and D. Vargas, PDA J.

Pharm. Sci. Technol. 60 (4), 248–253

(2006). ◆

Silicone tubing has a long history of safe

use for a broad range of bioprocess fluids, and

provides high temperature and

gamma radiation stability.

Page 25: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 23

Retrospective

Throughout BioPharm International’s 25th anniversary year, we will be looking back at articles published in the first volume of the journal. This month, Jerold Martin of Pall Life Sciences takes a look at protein recovery through direct-flow microporous membrane filters over the past 25 years.

Protein binding to membrane filters was a “hot

topic” back in 1988. While biologicals were

routinely sterile filtered through Nylon, cel-

lulose ester, or PVDF membranes without notable

losses, concerns were raised over reports of reduced

cell culture response in multi-well plates filled with

sequentially filter-sterilized culture media using

25-mm membrane syringe filters. Although inhi-

bition from initial filter leachables was not ruled

out, the primary cause of poor cell growth in initial

filled wells was attributed to binding and depletion

of nutrient protein growth factors. Consequently,

process developers began focusing on initial bind-

ing of proteins to membranes in large-scale protein

filtration processes. Bench-scale data were often

misleading because it was frequently misunder-

stood that protein binding to microporous mem-

branes is non-specific and will vary with protein

type, concentration, pH, and other formulation

components. Membrane filters do not have a finite

“protein binding capacity” independent of formu-

lation and process conditions.

Some membranes that incorporate surfactant

wetting agents and are not preflushed showed

low initial protein binding that can increase in

later throughputs when the wetting agent (an ini-

tial leachable) is washed out. Other membranes

showed relatively high protein binding during

initial throughputs, but soon saturated so that the

impact on total protein recovery in bulk fills was

negligible. The value of even microgram quantities

of proteins potentially lost to binding on a mem-

brane became significant as costly biotech protein

drugs at low concentration were introduced.

The 1988 paper, “Protein Recovery from

Effluents of Microporous Membranes,” cowrit-

ten with Richard L. Manteuffel, explained the

dynamics of protein binding to membrane filters

and directed process developers to more appropri-

ate evaluation protocols. Many researchers in the

1980s were evaluating protein binding by making

up a single concentration of a model protein in a

model buffer, then assessing loss after only a few

mLs of throughput through a small membrane

disc and applying their conclusions to selection

of process scale filters. While this protocol will

effectively distinguish “high” from “low” pro-

tein binding membranes, it is misleading because

binding can vary significantly for other proteins

at other concentrations, or at different pHs in dif-

ferent buffers. The protocol missed that binding is

transient once the membrane saturates, typically

within 10–20 mL through a 47-mm disc.

Today, the dynamics of protein binding are

better understood, and saturation dynamics are

routinely measured for specific target protein for-

mulations. New membrane filters used today for

process-scale protein filtration, both for serum-

free culture media and dilute-protein biotherapeu-

tics, feature extremely low non-specific protein

binding surfaces. Most of these surfaces, either

on polyvinylidene fluoride or polyethersulfone

membranes, are covalently grafted from a mixture

of acrylate monomers to

form a hydrophilic, non-

ionic polyacrylate surface

on the internal pores of

the membrane. These

surfaces, comparable to

size exclusion chroma-

tography media and low

protein binding contact

lens materials, serve to

minimize non-specific

protein binding to mem-

branes even on initial

throughputs, which can

be important when fill-

ing containers directly

through membrane filters

without batch pooling. ◆

View the 1988 article, “Protein Recovery from Effluents of Microporous Membranes“

by Jerold M. Martin and Richard L. Manteuffel, at

BioPharmInternational.com/Retrospectives.

A 25-Year Retrospective on Protein Recovery through Membrane Filters

Page 26: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

24 BioPharm International www.biopharminternational.com November 2012

Co

ver

co

urt

esy:

Photo

dis

c/Thin

ksto

ck

Biosimilars

The European Union has reached

an important stage in its efforts

to make the region a biosimilars

production center. The London-

based European Medicines Agency (EMA),

which licenses EU pharmaceuticals, has

finalized a guideline on biosimilar mono-

clonal antibodies (mAbs) and is drafting

another one on the key issue of quality of

biosimilars (1, 2).

To supplement an eight-year-old

basic regulation laying down the gen-

eral principles behind the introduction

of biosimilars, EMA has issued a series of

product-specific guidelines. These cover

biosimilars for insulins, somatropins, gran-

ulocyte-colony stimulating factor (G-CSF),

epoetins, low-molecular weight heparin,

and interferon-α (3). However, the guide-

line on mAbs is seen as technically the

most challenging because of the complex-

ity of the drugs in terms of their biological

structure, range of clinical indications, and

potential for unwelcome adverse effects,

particularly immunogenicity.

The mAb guideline was published

at the same time as a separate one on

immunogenicity in mAbs, which the

EMA points out are associated with

unwanted immunogenicity (4). Because

the mAb guideline concentrates pri-

marily on safety and efficacy, quality

matters are being covered by the gen-

eral quality guideline for biosimilars,

currently in the consultation stage.

“The finalization of the EU mAbs

guideline can be regarded as another

major milestone in the scientific frame-

work for biosimilars,” says Suzanne Kox,

senior director of scientific affairs, at the

European Generic Medicines Association

(EGA), which is in Brussels.

EU Sets Guidelines for Biosimilar Monoclonal Antibodies

Sean Milmo

The European Medicines

Agency has added

granularity to its biosimilars

approval pathway by releasing a

guideline on biosimilar monoclonal antibodies

(mAbs)

Sean Milmo is a freelance writer based

in Essex, UK, [email protected].

Page 27: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 25

Biosimilars

THE HIGH COST OF DEVELOPMENTThe EMA, unlike with nationally-

approved generic drugs, is respon-

sible for licensing biosimilars and has

so far authorized 14—two somatro-

pins, five epoetins, and seven G-CSF

filgrastims. The publication of the

mAb guideline is expected to trigger

a sizeable number of mAb biosimi-

lars applications. As with previous

guidelines, however, the one on

mAbs confirms the expectation that

biosimilars production and develop-

ment will be an expensive business

because of the high cost of manufac-

turing facilities and of conducting

nonclinical and clinical trials.

“To get started in the biosimi-

lars business, a lot of investment is

required in biopharmaceutical pro-

duction facilities—probably around

€300–400 million ($390–520 mil-

lion),” said Andreas Barner, chair-

man of Boehringer Ingelheim, at an

R&D conference at Ludwigshafen,

Germany, earlier this year. “Even

more important will be the high

cost of developing biosimilars

because, for the first time, non-

original medicines will have to be

supported by large-scale clinical tri-

als,” he continued. “The sector will

suit companies which already have

biopharmaceutical plants and have

experience of bringing original

compounds to the market.”

In the EU, the cost of regula-

tory compliance for biosimilars

will be pushed up even further by

the implementation of the EU’s

new rules on pharmacovigilance,

which stipulate additional post-

launch monitoring of biological

drugs, particularly biosimilars,

and mAbs.

“[Pharmacovigilance] is particu-

larly important with monoclonal

antibodies as these are complex

molecules with complex safety

profiles,” says Alan Morrison,

chairman of the regulatory affairs

advisor committee of the UK

Bioindustry Association (BIA).

BIOSIMILAR MAB DEVELOPMENT CLARIFIEDThe mAb guideline’s overall aim is

to lay down general principles for

nonclinical and clinical studies on

any potential differences between a

biosimilar mAb and a reference mAb

and how much these differences may

amount to a dissimilarity between

the two products. The guideline

recommends a step-wise approach,

with both in vitro and in vivo studies

being conducted on a case-by-case

basis. For example, comparative in

vitro studies would be conducted first

to assess differences in binding or

function, with the necessity for addi-

tional steps involving in vivo work

being determined by the need for

additional information.

Extrapolation of clinical efficacy

Begin an Exciting Web Experience at

www.BioPharmInternational.com

Learn the latest

bioprocessing methods,

regulatory trends, and

business best practices

from authoritative industry

experts—along with

the industry news and

information you need.

EXPLORE WITH EASE THE MOST VALUABLE INDUSTRY INFORMATION.

www.BioPharmInternational.com

Page 28: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

26 BioPharm International www.biopharminternational.com November 2012

Biosimilars

and safety data to other indications

of the biosimilar, based on the over-

all evidence from the comparability

exercise, would be acceptable, accord-

ing to the guideline.

In its draft guideline on quality of

biosimilars, EMA states that, “it is not

expected that all quality attributes

(between the biosimilar and the ref-

erence product) will be identical and

minor differences may be acceptable,

if appropriately justified” (2). In the

mAb guideline, the agency points

out that assays have been developed

in recent years allowing more in-

depth characterization of complex

proteins both at the physicochemical

and functional levels. These assays

have enabled more effective assess-

ment of minor quality differences in

manufacturing processes for mAbs.

Nonetheless, EMA warns that “in the

current state of knowledge it may be

difficult to interpret the relevance

of minor quality differences” when

comparing a biosimilar with a refer-

ence mAb (1) .

PHARMACOVIGILANCE REQUIREMENTSRegulators expect that during

the postauthorization pharma-

covigilance stage, unpredicted

adverse results from differences in

manufacturing process could be

detected. Incorrect extrapolations

from nonclinical or clinical data

during comparability assessments

could also be revealed.

The EU’s pharmacovigilance leg-

islation, approved in 2010, simplifies

reporting of adverse drug reactions

and the submission of periodic safety

update reports (PSURs), data from

both of which will be held centrally.

Postauthorization safety and

efficacy studies can be requested

from drug-licence holders by the

authorities. Applicants for market-

ing approvals of medicines will

have to submit postauthorization

risk-management plans.

For biosimilars, particularly

mAbs, pharmacovigilance require-

ments will be tougher than for

conventional medicines. “The

pharmacovigilance system is an

essential part of monitoring biosim-

ilars after approval,” says Morrison.

“[The legislation] provides for addi-

tional monitoring for biological

products, including biosimilars.”

The mAb guideline states that

applicants for marketing autho-

rization of their mAbs or other

biosimilars will have to provide a

“comprehensive concept” of how fur-

ther postauthorization safety stud-

ies will be carried out. The safety

plan will cover safety claims based

on extrapolations of efficacy and

safety data during the comparability

exercise and occurrences of rare and

particularly serious adverse effects.In

addition, because adverse reactions to

biosimilars could be due to defects in

manufacturing processes, the prod-

ucts should be clearly identifiable by

name and batch number, according

to the mAb guideline.

A GLOBAL ENTERPRISEBecause of the high expenditure nec-

essary on manufacturing facilities,

development, and pharmacovigi-

lance activities, biosimilar produc-

ers are looking for ways of curbing

costs. Merck Serono and Dr. Reddy’s

are among pharmaceutical compa-

nies that have formed an alliance in

the production and development of

biosimilars, mainly mAbs, to spread

the risk. DSM of the Netherlands

and Crucell, a Dutch subsidiary of

Johnson & Johnson, have reduced

the scope of Percivia, their US-based

joint venture in biosimilars by end-

ing in-house product development.

“In order to reduce the overall

development costs, we need in the

EU and in the US a scientific frame-

work which allows global devel-

opment,” says Kox. The European

Commission has appeared to be

acknowledging the need for global

development of biosimilars when

it recently announced it would

accept applications for approvals of

biosimilars containing data from

reference products that come from

outside the EU.

“[With this announcement] a new

regulatory paradigm is born, which

is a great achievement after years of

discussion,” says Kox. “The issue of

the reference product is not the only

one to achieve global development.

But for companies to be able to use

data from tests from reference prod-

ucts that are not sourced from the EU

constitutes a major and far-reaching

regulatory breakthrough.”

The European Commission has

yet to finalize details on how a com-

parability exercise with a reference

product outside the EU would work.

But the decision does seem to show

that the EU is intent on continuing

to take a lead on biosimilar regula-

tory matters without waiting for US

guidance on the development of

biosimilars to be formally adopted.

FDA issued three draft guidance

documents earlier this year to assist

companies in developing biosimi-

lars for the US market (5–7). Under

the Patient Protection and Affordable

Care Act of 2010, biosimilars are due

to be allowed to be marketed in the

country by 2014.

REFERENCES 1. EMA, Guideline on Similar Biological

Medicinal Products Containing Monoclonal

Antibodies—Non-Clinical and Clinical

Issues (London, June, 2012).

2. EMA, Guideline on Similar Biological

Medicinal Products Containing

Biotechnology-Derived Proteins as Active

Substance: Quality Issues, revision 1

(London, May 2012).

3. EMA, “Multidisciplinary: Biosimilar,”

www.ema.europa.eu/ema/index.

jsp?curl=pages/regulation/

general/general_content_000408.

jsp&mid=WC0b01ac058002958c,

accessed Oct. 13, 2012.

4. EMA, Guideline on Immunogenicity Assess-

ment of Monoclonal Antibodies Intended for

In Vivo Clinical Use (London, June, 2012).

5. FDA, Guidance for Industry: Q & As

Regarding Implementation of the BPCI Act

of 2009 (Rockville, MD, Feb. 2012).

6. FDA, Scientific Considerations in

Demonstrating Biosimilarity to a Reference

Product (Rockville, MD, Feb. 2012).

7. FDA, Quality Considerations in

Demonstrating Biosimilarity to a

Reference Protein Product (Rockville,

MD, Feb. 2012). ◆

Page 29: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

EVENT OVERVIEW:

Learn about the latest trends in global laboratory instrument

compliance. The speaker will address laboratory best practices

for meeting regulatory and compendial requirements, FDA’s

expectations on data integrity, operational qualification report

generation, and the GAMP Good Practice Guide for Validation of

Computerized Systems. In addition, an update will be provided

on USP Informational Chapter <1058> on analytical instrument

qualification.

ON-DEMAND WEBCAST

Register free at www.biopharminternational.com/labcompliance

Top Trends in Laboratory Instrument Compliance

Who Should Attend:

This webcast would be of interest to:

■ Compliance Directors and

Managers

■ Laboratory Directors and

Managers

■ Regulatory Affairs personnel

■ QA/QC Managers

Key Learning Objectives:

■ Hear the latest trends in

laboratory instrument

compliance processes

■ Learn about recent regulatory

and compendial updates

affecting laboratory instruments

and their implications

■ Gain insight from peers on

best practices for laboratory

compliance, including for high

performing labs

For questions, contact Sara Barschdorf at [email protected]

Presenter:

Paul Smith

EMEA and India Compliance Program Manager

Agilent Technologies

Moderator:

Angie Drakulich

Editorial Director

BioPharm International

Presented by Sponsored by

Page 30: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

28 BioPharm International www.biopharminternational.com November 2012

Upstream Processing

Cell-based therapies are gear-

ing up to have an extensive

impact on the healthcare

f ield in the coming years.

They show great promise in the treat-

ment of diseases ranging from various

types of cancer to chronic conditions

such as heart disease and diabetes.

The overall market for regenerative

medicine has been predicted to reach

as much as $20 bi l l ion by 2025,

according to research from Scientia

Advisors, and if this growth is to

come about, there must be reliable,

efficient methods for scaling up the

production of the stem cells that are

used in many of these therapies (1).

In initial phases of development,

the focus is on f inding a way to

obtain the desired cells, with cost

being less of a driver. However, on a

larger scale, cost becomes a signifi-

cant issue, and creates the need for

alternative manufacturing technology

to be implemented. It is also impor-

tant that emphasis remains on the

safety and reproducibility of the pro-

cess, as well as adherence to GMP

standards.

When growing adherent stem cells,

a surface must be provided. In the

laboratory, this is relatively simple,

and there are several systems avail-

able. F lasks and mult iplate stacks

allow adherent stem cells to grow suc-

cessfully on a two-dimensional (2D)

surface. However, problems appear

when the product moves into the

later stages of clinical trials and larger

numbers of cells are required. It must

Considerations for Scale-Up of Stem-Cell Cultures

Matthieu Egloff and

Jose Castillo

Scaling up stem-cell cultures requires careful

consideration of the bioreactor

design.

Matthieu Egloff is product manager

and Jose Castillo is global director of cell

culture at ATMI LifeSciences.

Ph

oto

Cre

dit

: iS

toc

kp

ho

to/T

hin

kS

toc

k im

ag

es

Page 31: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

200 Trays for Cell Expansion...

Xpansion™: the bioreactor solution to scale-up cell production without changing the environment for the cells.

The Xpansion™ bioreactor is a compact single-use and fully controlled system designed for adherent fragile cell culture like stem cells. The Xpansion design is based on stacked plates, made from the same plastic material as multiple-tray stacks. Keeping the same environment for the cells enables easy and straightforward transfer from multiple-tray stacks process to Xpansion bioreactor. With a capacity of 122.000 cm2, one single Xpansion bioreactor can replace up to 200 traditional stacked trays, providing an efficient solution for the production of large amount of cells.

ēƎ�+),��0Ǝ/%*#(!Ĩ1/!Ǝ�(+/! Ǝ/5/0!)

ēƎù�Ǝ,(�0!/Ǝ/1."��!/

Increase efficiency in cell therapy manufacturing.

© 2012 ATMI, Inc. All rights reserved. ATMI, the ATMI logo and Xpansion are trademarks or registered trademarks of Advanced Technology Materials, Inc. in the U.S., other countries, or both.

...Now in One Compact Closed System!

[email protected]

ēƎ�1((Ǝ�+*0.+(Ǝ+2!.Ǝ,Ǝ�* Ǝ��

ēƎ�!((Ǝ !*/%05Ǝ)+*%0+.%*#

Page 32: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

30 BioPharm International www.biopharminternational.com November 2012

Upstream Processing

be noted that there is a differ-

ence between the processes for

autologous and allogeneic thera-

pies. For an autologous therapy—

one based on the patient’s own

stem cells—the batches remain

the same size, but many more

batches are required, so scale-

out is needed. However, for an

a l logeneic therapy, where a l l

patients receive the same cells,

the batches become much larger,

and the process must be scaled

up, so a much larger surface area

is required.

SCALING UP ADHERENT STEM-CELL PRODUCTIONThere are currently three main

options available for scaling up

adherent cell production. These

include keeping a 2D surface

with some form of plate-based

system, using microcarriers in

stirred tank bioreactors, where

the cells grow on the surface of

the microbeads, or using some

form of three-dimensional (3D)

technology where a sophisticated

scaffold is used to hold the cells,

such as a microfiber or a hollow

fiber.

There are two main criter ia

that must be considered when

selecting a system for stem-cell

bioprocessing and recovery. The

growth, integrity, and quality of

the cells must be preserved and

controlled, and it must be pos-

sible to harvest and recover the

cells smoothly.

For scale-up, it is not as simple

as merely providing a larger sur-

face area. Fragile, adherent stem

cells are sensitive to the niche

microenvironment, which can

affect the way they differentiate.

For cell-therapy applications, it

is vital to prevent the cells from

making unwanted differentia-

tions.

Changing the niche environ-

ment in which they grow by

altering the surface can have a

large effect on the cell behav-

ior. Stem-cell culture and differ-

entiation are sensitive to many

different factors. These factors

include the physicochemica l

environment, pH, dissolved oxy-

gen, shear stress, and the con-

centration of metabolites within

the culture. The surface compo-

sition and geometry affect the

cells’ connections and the extra-

cellular matrix. The cell density

can also have an effect.

A not he r p o i nt t hat mu s t

be considered is the ability to

recover the stem cells smoothly.

Most of the systems used to grow

cell lines were designed for the

production of vaccines or pro-

teins, with no need to harvest

the cells at the end of the pro-

cess.

Overa l l, sca le -up f rom the

R&D laboratories to an indus-

trial process requires the ability

to control the physicochemical

parameters, to minimize the

change in the cell surface, and to

monitor cell density. The follow-

ing sections describe three com-

mon scale-up platforms.

Plate-based technologies

The main feature of a plate-

based system, such as the ATMI

LifeSciences Integrity Xpansion

multiplate bioreactor, is that the

surface remains flat and is made

from hydrophilized polystyrene.

The plates are similar to those

on which stem cells are grown

on multiplate stacks in the labo-

ratory. This bioreactor offers a

compact version of a laboratory

multiplate stack and enables one

to replace multiple devices by a

single container. One Xpansion

200 plate of fers a sur face of

120,000 cm2 (equivalent to 20

standard stacks of 10 plates) in

a 60x35 cm area. In this model,

the bioreactor enables stem-cell

expansion in a closed system

with fewer operations needed.

The gas exchange is not between

the plates, but rather in a cen-

tral column with channels along

the plates through which the

medium circulates, consequently

reducing the footprint.

Temperature, pH, and d is -

solved oxygen levels are all care-

fully monitored and controlled.

Light microscopy is then used

to study the morphology of the

cells and enable cell density to

be calculated.

Harvesting the cells is also

straightforward. To remove them

from the plates, a solution of

an enzyme, such as trypsin, is

added to separate the cells from

the surface and prepare them for

concentration and washing.

Multiplate-based technologies

provide one solution for both

large autologous batches and

small, allogenic batches contain-

ing a few billions of cells per

batch.

Microcarrier systems

The most commonly used large-

scale technology involves micro-

carr iers. Here, the surface on

which the cells grow is in the

for m of m ic robeads , wh ich

are suspended in the culture

medium within a stirred tank

bioreactor. However, the envi-

ronment is modif ied, because

When a very large

number of cells are

required, microbeads

in suspension or

a high cell-density

bioreactor (i.e., fixed

bed) may be the

best option.

Page 33: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 31

Upstream Processing

continuous st i r r ing increases

the shear stress on the cells, and

this leaves no guarantee that

the morphology of the cells will

remain the same as when the

cells are grown on static surface,

such as plates.

Not all stirred tanks are suit-

able for use with microcarriers

because it is imperative that the

culture must be agitated very

smoothly and gently to ensure

that the mic robeads do not

break, and shear stress must be

minimized. To address this, a

few stirred tank bioreactors have

been designed with mixing tech-

nology that provides eff icient

mixing with minimal agitation

and lower shear stress.

One drawback of microbeads

is the seeding step. While they

have great potential at a large

scale, to be able to run a 200-L

bioreactor, it is necessary to have

gone through several scale-up

steps, with runs at 20 L and 50

L. Multiple scale-up steps add

to the complexity of the seed-

ing and passages steps. Another

drawback of the stirred tank is

a technological one: harvesting

the cells. A sophisticated and

gentle purification step becomes

essential to separate the cells

from the beads without damag-

ing or changing them.

Microbeads represent a solu-

tion for large-scale production,

but process deve lopment i s

highly complex and takes time,

and cell observation is complex.

The microcarrier technology, on

the other hand, optimizes capac-

ity and is promising for large

allogenic batches that have more

than hundreds of billion cells

per batch.

Packed-bed bioreactors

Another alternative for adher-

ent cell growth is a packed-bed

bioreactor. This type of bioreac-

tor has a 3D scaffold structure

based on a specific material such

as nonwoven microfibers, a bio-

polymer, or a hydrogel, and the

cells are entrapped in this fixed

bed. A drawback is the difficulty

of observing the cells as they

grow. This technology provides

a good environment for the cells

to grow in, but because of its 3D

nature, there is an even greater

possibility that the morphology

of the stem cells will be differ-

ent and as a result, a significant

amount of validation is required

to ensure the final production

process is both robust and well

def ined. A fur ther drawback

of this technology comes with

the harvesting process. During

harvesting, the cells need to be

removed from the scaffold.

It has been proven that the

cells grow in hollow fiber-based

sy s tems because t hey g row

within the fibers, limiting the

shear stress. It is notable that

these fibers are limited in terms

of scale, and the pH and dis-

solved oxygen within them may

not be controlled. However, if

the cells are particularly sensi-

tive to shear stress, packed-bed

bioreactors offer advantages over

stirred tanks.

The common drawback across

all of these systems is that the

surface has changed from 2D to

3D, which may lead to different

behavior in the cells. However,

biopolymer, hydrogel, and inno-

vative scaffolds may a means of

If the cells are

particularly sensitive

to shear stress,

packed-bed bioreactors

offer advantages over

stirred tanks.Together, we can

produce mycoplasma-

free cell culture

media...............faster

www.parker.com/processfiltration

Europe: phone +44 (0)191 4105121email: [email protected]

North America: toll free: +1 877 784 2234 email: [email protected]

The new PROPOR MR mycoplasma retentive filter from Parker domnick hunter delivers industry leading flow rates without compromising on mycoplasma retention assurance.

Find out more at: www.parker.com/mycoplasmafree

Validated mycoplasma removal

Reduced filtration costs

Faster batchprocessing

Typical LRV >10 for

Acholeplasma laidlawii

Industry leading

flow rates

Integral prefilter layer

for increased capacity

Page 34: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

32 BioPharm International www.biopharminternational.com November 2012

Upstream Processing

solving the drawback of alloge-

neic therapy and large-scale stem

cell expansion.

CHOOSING THE BEST OPTIONIn the early stages of develop-

ment, laboratory technolog y

such as cell stacks or f lasks is

perfectly adequate to produce

a sufficient amount of cells for

preclinical and clinical studies.

But once Phase I and II tr ials

have succeeded and the prod-

uct moves down the develop-

ment pipeline, these methods are

impractical, and some form of

scale up or scale out is necessary.

This process is further compli-

cated by the fact that GMP stan-

dards also require that safety and

process reproducibility be con-

sidered and documented.

When choosing the best bio-

reactor, a number of questions

need to be asked. These ques-

tions include looking at whether

shear stress, pH, and dissolved

oxygen levels need to be con-

trolled. This is mandatory with

fragile cells such as stem cells.

A nother impor tant quest ion

is related to the scale of batch.

When a very large number of

cells is required (i.e., hundreds of

billions cells per batch), micro-

beads in suspension or a high-

cell-density bioreactor (i.e., fixed

bed) may be the best option. On

a more limited scale, cell stacks

can be used if the scale remains

extremely smal l—mil l ions of

cel ls. A mult iplate bioreactor

will probably be more appropri-

ate for mid-scale production (i.e.,

hundreds of millions of cells per

batch).

Overa l l, the sca le -up f rom

R&D to industr ial production

must preserve the integrity and

quality of the cells. It is impor-

tant to be able to control the

physicochemica l parameters,

minimize change in the cel l

surface, and monitor cell den-

sity. The process must meet GMP

standards within a closed sys-

tem, be reproducible, and require

minimal operator intervention.

For an autologous therapy,

the most practical option may

well be a 2D multiplate system

such as the Xpansion bioreactor

which has a much smaller foot-

print and operator requirement

than laboratory-scale devices.

The biggest advantage of this

bioreactor is that the microenvi-

ronment in which the cells grow

remains close to the environ-

ment in the cell stack or f lask,

and therefore, the way in which

the cells behave is much more

likely to remain the same as in

small-scale culture. As such, it

requires less process develop-

ment effort and decreases risk,

because of the similarity to labo-

ratory-scale systems. Automation

of the system can be a solution

to support scale out, and can

enable running several cultures

in parallel to supply large vol-

umes for commercialization.

For an a l logeneic therapy,

where the volume requirements

may be much larger, switching

over to using microcarriers in

a stirred tank bioreactor could,

in the long run, be the best

solution. An intensive process-

development program would

be essential to ensure that the

stem cells that are grown remain

substantially the same as those

made in the laboratory, and that

neither the changed microen-

vironment nor the harvesting

process affect the final product.

Packed-bed technologies would

be even more efficient, but inno-

vat ive and spec i f ic sca f folds

must be developed if they are

to become a practical solution.

A mixed scale-up and scale-out

process using Xpansion might

offer a route to push forward

cl inical development and the

early commercial izat ion scale

while more efficient large-scale

technologies are being investi-

gated.

THE FUTUREWhen working on a larger scale,

microcarriers in a stirred-tank

bioreactor and packed-bed biore-

actors can enable larger batches

to be made, but may require

much more development work

to ensure the cells grow with the

correct morphology. Multiplate

bioreactors address this chal-

lenge by mimicking the labora-

tory-scale equipment in which

the cells are initially developed.

In the long r un, indust r y

needs to develop a new solu-

tion that simplifies the scale-up

process. The perfect technology

w i l l minimize the t ime and

energy required in the devel-

opment process, while ensur-

ing the production of stem cells

with the correct morphology in

reproducible batches. The mul-

tiplate design is practical in the

earlier stages of development as

an enabling technology for the

industrialization of cell therapy

process. Future technological

development will ultimately be

required to support and sustain

the mass commercialization of

stem-cell therapies. ◆

REFERENCE 1. B. Nafzinger, “Regenerative Medicine

to Be a $20 Billion Industry by 2025,”

April, 2010, www.dotmed.com/news/

story/12382?p_begin=0, accessed

Oct. 22, 2012.

Page 35: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

*This offer is only available to end-users. If you are a consultant or solution provider

please call us on +44(0)20 7202 7690 for our commercial opportunities”

Researched & Produced by:

Key topics include:t� .BYJNJTJOH�ESVH�JOWFTUNFOUT���%FWFMPQJOH�

CVTJOFTT�GSBNFXPSLT�GPS�UIF�TVDDFTTGVM��DP�DSFBUJPO�PG�WBMVF

%S�"OEZ�1BSTPOT �

VP Preclinical Development, GSK

t� 4VDDFTTGVMMZ�JEFOUJGZJOH�IVNBO�ESVH�NFUBCPMJUFT�GPS�UPYJDJUZ�UFTUJOH

*BO�8JMTPO �

Senior Principal Scientist, "TUSB;FOFDB

t� 6TJOH�USBOTMBUJPOBM�NFEJDJOFT�JO�FBSMZ�ESVH�EFWFMPQNFOU

-VDFUUF�%PFTTFHHFS �

Global Head of Translational Safety Medicine, 3PDIF

To find out more visit www.eddsummit.com or call +44(0)20 7202 7690

Save £400

on your registration

7JTJU��XXX�FEETVNNJU�DPN�PGGFS

Page 36: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

34 BioPharm International www.biopharminternational.com November 2012

Peer-Reviewed: Cell Harvesting

The first step in the recovery

of a secreted product from

a mammalian cell culture is

to separate the cells and cell

debris from the product in the super-

natant of the cell culture. A disc-stack

centrifuge or tangential flow filtra-

tion and microfiltration (TFF–MF) is,

generally, used to separate cells as a

primary recovery step, followed by

depth filtration as a secondary step

to remove remaining cell debris and

other impurities before the product in

the clarified liquid (centrate) can be

loaded into a chromatography column

(1–3). More information on these pri-

mary technologies can be found in a

review paper (4).

Most conventional cell harvesting

technologies are not single-use sys-

tems and, therefore, require extensive

cleaning and sterilization between

batches during production. Single-use

technologies have been widely used

in the biotechnology industry due to

their distinct advantages. Within the

past decade, a variety of cell-culture

processes has adapted single-use sys-

tems, ranging from the use of WAVE

technology for processes up to the 500

ABSTRACTThis article discusses the evaluation of a novel single-use fluidized bed centrifuge (FBC) for harvesting of antibodies. An FBC that contains four single-use 100-mL chambers was used to harvest Chinese Hamster Ovary (CHO) cell cultures. Optimal operating parameters were defined by performing preliminary studies to determine the maximum chamber capacity and the feed flow rates into the centrifuge. Results of the preliminary studies showed the maximum capacity was approximately 10x109 cells/chamber, and the initial and process feed flow rates used for the studies were 80 and 140 mL/min/chamber, respectively. Five simulated cell-harvesting runs followed the preliminary studies. Three of the five simulated runs utilized healthy cell cultures with viabilities > 90%, and the remaining two runs were with cell cultures with viabilities < 50%. Results showed that the FBC was efficient in separating cells from the product, with low cell density and turbidity detected in the centrate. Average clarification efficiencies were between 88–93% without the use of depth filtration post-clarification. There was no increase in lactate dehydrogenase (LDH) and residual DNA levels indicating that minimal amount of shear stress was induced by the centrifugation. The results, therefore, suggest that the FBC is a promising alternative for cell-harvesting applications.

Evaluation of Single-Use Fluidized Bed Centrifuge System for Mammalian

Cell HarvestingHsu-Feng Ko and Ravi Bhatia

Hsu-Feng Ko is a research scientist and Ravi Bhatia is associate director, both at Janssen

Research & Development, Spring House, PA.

[email protected]

PEER-REVIEWED

Article submitted: Oct. 27, 2011

Article accepted: Mar. 13, 2012

Ima

ge c

ou

rte

sy o

f kS

ep S

yste

ms

Page 37: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 35

Peer-Reviewed: Cell Harvesting

L scale as well as single-use stirred-tank bio-

reactor (SUBs) technology up to the 2000

L scale as the production bioreactor ves-

sel for therapeutic protein production (5,

6). Several single-use cell harvesting tech-

nologies have also been utilized, including

but not limited to the Pod Filter system

from Millipore, TFF bioprocess systems from

SciLog, and Sartoclear depth filter systems

from Sartorius Stedim.

In the past two years, single-use cen-

trifuge technologies have also emerged

as a potential a lternative for pr imary

recovery processes. The Unifuge, made by

Pneumatic Scale Angelus, is a fully auto-

mated centrifuge system that uses a sin-

gle-use insert inside the centrifuge bowl.

The kSep technology, a FBC made by kSep

Systems Inc., uses balancing centrifugal

and fluid flow forces to capture cells in up

to four single-use chambers. Both tech-

nologies involve retaining cells inside the

centrifuge while the centrate that contains

the product is continuously separated and

discharged. Because of the FBC’s advan-

tages (e.g., washing capability, low shear,

and continuous mode of operation), the

authors selected the FBC for the evalua-

tion of mammalian cell harvesting. The

FBC and its single-use set are shown in

Figure 1.

Compared with cell harvesting alterna-

tives that rely on the use of conventional

centrifuges such as disc-stack or filtration

devices to separate cells from the superna-

tant, the FBC has several advantages over

both conventional methods. First, the FBC

eliminates the need for cleaning and ster-

ilizing cycle validations. Second, there is

no risk for cross-contamination between

batches because the product-contact com-

ponents are single-use only. Third, there

is lower shear stress induced by the FBC

during operation compared with con-

ventional centrifugation and filtration.

Because of the establishment of the fluid-

ized bed during operation, FBC’s g-force

does not result in cell lysis because cells

are not packed against the centrifuge wall.

Finally, the washing option available with

the FBC provides maximal recovery of the

product without diluting the centrate. In

addition to these advantages, historical

clarif ication efficiency data from disc-

stacks have been shown to be comparable

with the expected clarification efficiency

performance of the FBC (7).

The FBC is based on a balancing act of two

counteracting forces within the system, the

centrifugal force (Fg) versus the fluid flow

(VQ). The physics of the FBC follows Stokes’

Law for spherical particles in a continuous

fluid (Equation 1), or in the case of bioprocess-

ing, a cell suspended in a medium:

= V2 2D

9μ 4p f

s g (Eq. 1)

where Vs is the settling velocity of the

particle, ρp is the density of the particle, ρf

is the density of the fluid, μ is the dynamic FIG

UR

E 1

CO

UR

TE

SY

OF

kS

EP

SY

ST

EM

S. F

IGU

RE

S 2

–6

AR

E C

OU

RT

ES

Y O

F T

HE

AU

TH

OR

S

ChamberFluidized Cell Bed

Flu

id fl

ow

fo

rce

Ce

ntr

ifu

ga

l fo

rce

Spent Media Outlet

Cell CultureInlet

Shaft

Rotor

Figure 2: FBC schematic demonstrating principles of FBC’s

fluidized bed.

Figure 1: FBC (left) and single-use set (right) loaded into the centrifuge

rotor (Image courtesy of kSep Systems).

Page 38: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

36 BioPharm International www.biopharminternational.com November 2012

Peer-Reviewed: Cell Harvesting

viscosity of the fluid, D is the diameter of the

particle, and g is the gravitational acceleration

of the particle. The centrifugal force (Fc) and

fluid flow velocity (VQ) are defined in the fol-

lowing equations (Equations 2 and 3):

= F 2Rc (Eq. 2)

VQ

AQ = (Eq. 3)

where ω is the angular velocity, R is the

radius, Q is the flow rate, and A is the cross-

sectional area of the chamber. When the

settling velocity (VS) due to the centrifu-

gal force is balanced against the fluid flow

velocity (VQ), a fluidized cell bed is created

within the chambers while the supernatant

is discharged from the chamber as the cen-

trate. Figure 2 shows a schematic of the inner

workings of the FBC.

The overall preparation time for the FBC is

relatively short compared to the cycle times

for cleaning and sterilizing stainless-steel

equipment. A single-use set that contains

four interconnected chambers and the valve

set are first inserted into the FBC prior to an

operation. After loading the single-use set

into the FBC, individual tubing ends from

the single-use set can be sterile-connected

to respective vessels such as the bioreactor,

buffer, centrate vessel for harvested prod-

uct, and waste container. Pinch valves and

bubble sensors are built-in as part of the FBC

and used to automate the cell-harvesting pro-

cess. The built-in human-machine interface

(HMI) screen on the FBC allows run param-

eters such as centrifuge speed, flow rates,

and harvest volumes to be set in individual

recipes. Cell-harvesting runs can be nearly

fully automatic once the recipes are config-

ured, but manual controls are also available

if required.

Figure 3 shows the process flow diagram

of a typical harvest clarification recipe. Step

one in the process is to prime the single-use

set with a buffer. All tubing and chambers

are filled with the buffer to displace air from

the system. Step two in the recipe directs the

system to displace the buffer initially in the

single-use chamber(s) into the waste vessel.

In doing so, the buffer is not introduced into

the centrate vessel and will not dilute the

product. Harvest clarification begins in step

three, where cells and cell debris are retained

in the chamber(s) and the clarified liquid is

separated into the centrate vessel. When the

chamber(s) are nearly filled with cells, in step

four, the bioreactor feed temporarily pauses,

and the buffer is used to flush the chamber(s)

to recover the product that is still present

in the cell bed. In step five, after the wash,

pump directions are reversed to discard the

cell bed into the waste vessel. In the case

where the bioreactor volume is larger than

what all four single-use chambers can handle

in one batch, steps two through five can be

repeated in multiple cycles to finish process-

ing the full volume of the bioreactor. At the

end of the process, the system purges all

liquids from the single-use set and prompts

the operator to seal all tubing prior to the

disposal of the single-use set. The remainder

Step 1: Prime systemwith wash buffer

Step 2: Displace initial primebuffer (reduces product dilution)

Step 3: Start clarification, collectclarified media as harvest

Step 4: Wash cells with buffer(recovers product in chamber)

Step 5: Discard cell slurry byreversing pump direction

Repeat steps until bioreactor is empty,

then stop process, purge system and

seal tubing

Figure 3: Process flowchart for harvest clarification using the FBC.

Cell density in centrate

Elapsed time (min)

Cell d

en

sity

(x106 c

ells/

mL)

2.5

2

1.5

1

0.5

00 5 10 15 20 25 30 35 40 45 50 55

18

16

14

12

10

8

6

4

2

0

Estimated total cells in chamber

Est

imate

d c

ells

in c

ham

ber

(x10

9 c

ells)

Figure 4: Determination of maximum chamber capacity: cell density

and estimated total cells.

Page 39: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 37

Peer-Reviewed: Cell Harvesting

of this article discusses the results obtained

from the evaluation runs conducted using

the FBC for cell harvesting and clarification.

MATERIALS AND METHODSMaterials

All studies used CHO cells cultured in shake

flasks (Corning) followed by expansion into

WAVE bioreactor (GE Healthcare). Cells were

cultured in Janssen R&D’s proprietary, chem-

ically-defined medium. 1X phosphate-buff-

ered saline (PBS, Gibco) was used for priming

and rinsing during the runs.

The FBC (model kSep 400) and the single-

use sets were purchased from kSep Systems,

Inc. Cells were placed on an orbital shaker

(VWR International) set at 100 RPM during

operations to keep cells in suspension.

Cell culture

Because of the simulated nature of the study,

CHO cells were only cultured up to four days

and up to a cell density of approximately

5x106 cells/mL. To simulate cell cultures with

lower viabilities such as those found in 18–20

day cultures to harvest antibodies or thera-

peutic proteins, a second batch of cells were

intentionally starved/asphyxiated by turning

off nutrient and air supply after day four and

then mixed with healthy cultures to lower

the viability.

Determination of

maximum chamber capacity

The maximum number of cells each cham-

ber can hold is of great significance because,

in most applications, the total number of

cells from a bioreactor will exceed what the

four chambers can hold. The maximum

capacity can also vary from cell line to cell

line and is dependent on factors such as cell

size. It is, therefore, crucial to determine the

maximum chamber capacity at which break-

through of the cell bed will occur. For this

experiment, one chamber was used and the

centrifugal force was maintained at 1000g.

Cell suspensions were continuously pro-

cessed by the FBC at 140 mL/min/chamber

and samples were taken from the centrate

to examine the amount of cells escaping

from the chamber. Samples were taken until

the breakthrough threshold of the chamber

capacity had been reached. Cell count was

performed using Cedex cell counter (Roche).

Determination of initial feed flow rate

The initial flow rate when cell suspensions

are first introduced into the FBC was deter-

mined. It is hypothesized that prior to the

formation of the cell bed in the chambers,

the flow rate should be kept lower to mini-

mize potential escape of cells from the cham-

bers due to initial instability of the cell bed.

To test this hypothesis, 100 mL/min/cham-

ber and 80 mL/min/chamber were compared

in two separate runs. Two chambers were

used in each of the runs, and the centrifugal

force was maintained at 1000g. Cell counts

samples were taken and performed on the

Cedex to determine the cell density.

Determination of optimal

process feed flow rate

Once the fluidized bed is established in the

single-use chamber and is stable, the feed

flow rate can then be increased. Consecutive

runs were performed where process flow

rates were increased incrementally from 140

to 225 mL/min/chamber while maintain-

ing the centrifugal force at the maximum of

1000g. Cell counts samples were taken from

the centrate to determine the cell density.

One chamber was used in each of the runs.

Cell harvesting with FBC

Five total cell-harvesting runs were com-

pleted using the FBC. The first three runs

were conducted using CHO cell cultures with

> 90% viability, followed by two remaining

runs using CHO cell cultures with < 50% via-

bility. For all runs, the centrifugal force was

kept at the maximum g-force of 1000g. The

initial feed flow rate was 80 mL/min/cham-

Flowrate (mL/min/chamber)

Cell d

en

sity

(x106 c

ells/

mL)

30.0

25.0

20.0

15.0

10.0

5.0

0.0120 140 160 180 200 220 240

Viable cell density

Total cell density

Figure 5: Centrate cell density over time at various process flow rates.

Page 40: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

38 BioPharm International www.biopharminternational.com November 2012

Peer-Reviewed: Cell Harvesting

ber during initial establishment of the cell

bed, and increased to 140 mL/min/cham-

ber after 5 min into the process. Samples

were taken to measure cell density, turbidity

(NTU), LDH level, and residual DNA content.

Cell density and viability were determined

using the Cedex cell counter. Turbidity

was measured using the HACH 2100AN

Turbidimeter. Cell density and turbidity were

also used to calculate the FBC’s clarification

efficiency using Equations 4 and 5:

Clarification Efficiency(Cell Density)

1CD

� 100CD

Centrate

Bioreactor

= (Eq. 4)

1NTU

� 100NTU

Centrate

Bioreactor

Clarification Efficiency(NTU)

=

(Eq. 5)

where CDCentrate is the final cell density

in the final harvest vessel, CDBioreactor is

the starting cell density in the bioreactor,

NTUCentrate is the turbidity in the final har-

vest vessel, and NTUBioreactor is the starting

turbidity in the bioreactor.

LDH levels and residual DNA from samples

before and after the FBC process were used as

a measure of shear stress and cell lysis dur-

ing the process. LDH and residual DNA were

measured using Johnson & Johnson Vitros

Chemistry System DTSC Module and Applied

Biosystems Prism 7500 Sequence Detection

System, respectively. Antibody titer was also

determined using Agilent 1100 Series HPLC.

Theoretical calculations

to estimate cell harvesting process time

Finally, because the studies discussed in this

article were simulations on a small scale,

theoretical calculations were performed to

estimate the total process time required for

larger bioreactor scales. Calculations were

performed using both the kSep 400 and the

process scale version, kSep 4000, for 50-L,

25-0L, and 1000-L bioreactors, assuming all

four single-use chambers were run at 1000g

and at various feed-flow rates.

RESULTS AND DISCUSSIONDetermination of

maximum chamber capacity

In Figure 4, the measured cell density in the

centrate is displayed in blue, and the esti-

mated total cell number based on the flow

rate and time is displayed in red. The cell

densities in the centrate were initially low

until approximately 9–10 billion cells were

retained in the chamber. After this point,

cell density in the centrate became exponen-

tial, as the cell density began to approach the

cell density of the bioreactor (~2.6x106 cells/

mL). The study was completed before the

centrate cell density actually reached the cell

density of the bioreactor. Otherwise, it would

have been observed where the cells entering

the chamber would directly exit into the

centrate, and the cell density in the centrate

would be equal to the cell density of the bio-

reactor. Based on the results of this study, to

minimize the amount of cells from escaping

into the centrate, the maximum amount of

cells per chamber should be kept below 10 x

109. This limit was factored into the remain-

der of the studies.

Determination of initial feed flow rate

Based on cell count results from the Cedex

cell counter, when running the FBC at 100

mL/min/chamber initially, a sharp spike

(result not shown) was observed in the first

few minutes of the run. When the initial

flow rate was reduced to 80 mL/min/cham-

ber, the spike was not observed. Hence, a

slower flow rate of 80 mL/min/chamber was

more optimal and was used in remaining

studies during the formation of the cell bed.

Determination of optimal

process feed flow rate

Cell densities, both viable and total, that

correspond to the amount of cells in the

centrate at various flowrates are shown in

Figure 5. As the flow rates increased from

140 mL/min/chamber to 225 mL/min/cham-

ber, both viable and total cell density in

LDH

Levels

(U

)

60000

50000

40000

30000

20000

10000

0>90% Viable <50% Viable

Bioreactor LDH Level

Centrate LDH Level

Figure 6: LDH level comparison between bioreactor and centrate.

Page 41: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 39

Peer-Reviewed: Cell Harvesting

the centrate increased exponentially as the

centrifugal force alone became increasingly

insufficient to retain cells inside the cham-

ber. The trend exhibited in Figure 5 suggests

that the process feed flow rate can poten-

tially be set at 160 or 180 mL/min/chamber

with only minimal amount of cells lost into

the centrate. The authors elected to use 140

mL/min/chamber as the process feed flow

rate for all studies reported in this article

because this flow rate is adequate to accom-

plish all of the objectives in the studies.

Increasing the process feed flow rate up to

160 or 180 mL/min/chamber can be a part

of future studies if reducing process time

becomes critical.

Cell harvesting with FBC

Table I lists the pre-FBC (bioreactor) and post-

FBC (centrate) data from three cell-harvest-

ing runs with high cell viability (> 90%) and

two cell-harvesting runs with low cell viabil-

ity (< 50%).

Cell density data collected from the cen-

trate indicated that the FBC was efficient

in separating cells from the supernatant as

shown by the low cell counts. Starting with

cell densities of 2.3x106 cells/mL (Run #1),

5.4x106 cells/mL (Run #2), 4.8x106 cells/mL

(Run #3), 4.3x106 cells/mL (Run #4), and

4.0x106 cells/mL (Run #5) in the bioreactor,

none of the cell densities measured from

the centrate exceeded 0.215x106 cells/mL.

Comparing the cell counts in the final har-

vest vessels to the starting bioreactor cell

counts for each run, the efficiencies of cell

removal were in the range of 95.7–98.7% for

all runs.

Turbidity data of the centrate samples,

measured in NTU, also reflected a similar

outcome as the cell densities. It was shown

by the large reduction in NTUs that the FBC

was efficient in separating cells from the

supernatant. Starting with NTUs of 38.4

(Run #2), 33.3 (Run #3), 63.7 (Run #4), and

57.4 (Run #5) in the bioreactor, the FBC

effectively separated cells from the superna-

tant, resulting in significantly reduced NTUs

in the range of 2.47–6.91.

The clarification efficiencies for each of

the runs, based on NTU measurements, were

93.4%, 90.2%, 89.7%, and 88.2% for runs #2

through #5, respectively. Similar results on

clarification efficiencies have been reported

using the disc-stack centrifuge technology (7).

LDH levels measured pre- and post-FBC

are shown in Figure 6. The average LDH levels

for > 90% viability cultures were 3027±313

U and 3459±785 U for pre-FBC and post-

FBC samples, respectively, and the LDH

levels for the < 50% viability culture were

43,611±2782 U and 37,287±10,419 U for pre-

FBC and post-FBC samples, respectively. Post-

FBC LDH levels did not increase compared to

pre-FBC levels, a clear indication and confir-

mation that no cell lysis occurred during the

process. This is a unique advantage of using

the FBC, because cells are suspended in a flu-

Table I: Starting bioreactor and final centrate parameters for cell harvesting runs. LDH is lactate dehydrogenase. NTU is turbidity.

Parameters Run 1 Run 2 Run 3 Run 4 Run 5

Bioreactor batch size (L) 17.5 10.8 10.4 11.8 12.8

Bioreactor cell density (x106 cells/mL) 2.3 5.4 4.8 4.3 4.0

Bioreactor viability (%) 98.0 98.5 97.0 43.5 39.6

Bioreactor turbidity (NTU) - 38.4 33.3 63.7 57.4

Bioreactor LDH (U) - 2795 3259 43,831 43,392

Bioreactor residual DNA (mg) - 12.9 - 49.6 -

Centrate volume (L) 16.7 11.6 11.3 11.4 12.6

Centrate cell density (x106 cells/mL) 0.04 0.215 0.132 0.123 0.051

Centrate turbidity (NTU) - 2.47 3.20 6.58 6.91

Centrate LDH (U) - 4014 2904 29,920 44,655

Centrate residual DNA (mg) - 12.1 - 51.4 -

Clarification efficiency (%) - 93.4 90.2 89.7 88.2

Total processing time (min) 102 60 65 63 66

Processing rate (min/L processed) 5.8 5.6 6.3 5.3 5.2

Page 42: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

40 BioPharm International www.biopharminternational.com November 2012

Peer-Reviewed: Cell Harvesting

idized bed rather than having a high g-force

packing them against the centrifuge wall. A

similar outcome was confirmed by the analy-

sis of residual DNA content. Residual DNA

for the > 90% viability culture were 12.9

mg and 12.1 mg for pre-FBC and post-FBC

samples, respectively, and the residual DNA

for the < 50% viability culture were 49.6 mg

and 51.4 mg for pre-FBC and post-FBC sam-

ples, respectively, again showing minimal

cell lysis in the FBC.

Centrate samples from both Run #3 and

Run #5 were analyzed to determine the

antibody titers. Minimal antibody titer

loss or dilution was observed after process-

ing with FBC due to FBC’s efficient wash-

ing capabilities.

Theoretical calculations to

estimate cell harvesting process time

Table II shows the estimated time for a

typical cell harvesting process based on

theoretical calculations using both scales

of the FBCs. Assuming all four single-use

chambers are used at 1000g and 180 ml/

min/chamber, approximately 1.2 hr and

5.8 hr are required to harvest a 50-L and

250-L bioreactor, respectively, using kSep

400. The estimated times of 42 min and

2.8 hr are required at 1.5 L/min/chamber

to harvest a 250 L and 1000-L bioreactor,

respectively, using kSep 4000. Combined

with the advantages the FBC possesses over

other cell harvesting alternatives, the FBC

is emerging as a promising option for cell

harvesting.

CONCLUSIONThe single-use FBC was evaluated for cell-

harvesting applications. After determining

the maximum capacity of each single-use

chamber and the initial and processing flow

rates, five total cell-harvesting runs were

completed. Cell density, NTU, LDH levels,

and residual DNA content from the centrate

were collected for evaluating the system.

Low cell density and turbidity in the cen-

trate indicated that clarification efficiency

was high, and little to no change in the

LDH level tle cell lysis during processing.

Compared to disc-stack centrifuges and other

current standard technologies, it can be con-

cluded that the FBC system is an attractive

single-use alternative to current options for

cell harvesting.

ACKNOWLEDGMENTSThe authors would like to acknowledge the

following individuals for their contributions:

Sunil Mehta and Tod Herman for providing

their expertise on the FBC; Divya Harjani

and Nikhil Patel for maintaining CHO cell

cultures and assistance in carrying out some

of the FBC runs; Meredith Rice for her assis-

tance with the turbidimeter; and the Janssen

R&D Process Analytical Support group for

analyzing the residual DNA and antibody

titer samples.

REFERENCES 1. R. Kempken, A. Preissmann, and W. Berthold,

Biotechnol. Bioeng. 46 (2), 132–138 (1995).

2. R. Van Reis, L.C. Leonard, C.C. Hsu, and S.E.

Builder, Biotechnol Bioeng. 38 (4), 413–422

(1991).

3. Y. Yigzaw, R. Piper, M. Tran, and A.A. Shukla,

Biotechnol Prog. 22 (1), 288-296 (2006).

4. D.J. Roush and Y. Lu, Biotechnol Prog. 24, 488–

495 (2008).

5. V. Singh, Cytotechnology. 30 (1–3), 149–158

(1999).

6. R. Bhatia, C. Wood, N. Richardson, and S. Ozturk,

ACS National Meeting (San Francisco, CA, 2006).

7. M. Iammarino et al., Bioprocess International, 5

(10) 38–50 (2007). ◆

Table II: Theoretical calculations showing estimated total process time using fluidized bed centrifuges.

Bioreactor scale

Scale (capacity) Feed flowrate 50 L 250 L 1000 L

kSep400 (4x100mL)140 mL/min/chamber 1.5 hr 7.4 hr

180 mL/min/chamber 1.2 hr 5.8 hr

kSep4000 (4x1000mL)750 mL/min/chamber 17 min 1.4 hr 5.6 hr

1.5 L/min/chamber 8 min 42 min 2.8 hr

Page 43: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 41

Boot Camp: Tech Guide

Sve

ta D

em

ido

ff/G

ett

y I

ma

ge

s

In this third part of a series of primers

with training experts from the National

Inst itute for Bioprocessing Research

and Training (NIBRT), Dr. Jayne Telford,

Bioanalytical Training Lead, discusses the

basics of biopharmaceutical product char-

acterization. NIBRT provides training, edu-

cational, and research solutions for the

international bioprocessing industry in state-

of-the-art facilities. Located in South Dublin,

it is based on an innovative collaboration

between University College Dublin, Trinity

College Dublin, Dublin City University, and

the Institute of Technology Sligo.

DEFINING PRODUCT CHARACTERIZATIONBioPharm: The primary goal of product char-

acterization is to ensure a product’s safety,

purity, identity, and potency. The harmonized

guideline Q6B, Specifications: Test Procedures

and Acceptance Criteria for Biotechnological/

Biological Products, from the International

Conference on Harmonization (ICH) spe-

cifically calls for determination of physi-

cochemical properties, biological activity,

immunochemical propert ies, purity and

impurity. Can you go into more detail on why

these characteristics are crucial for overall

product and patient’s safety?

Telford: We are talking about protein-based

biotherapeutic drugs and their desired effect

in the patient, which occurs by interacting

with receptor proteins, with other proteins,

or with other targets, to enhance or inhibit

signaling or interactions in the body. For

proteins to have the desired effect, they must

have the correct sequence, the correct size,

the correct structure, and the correct post-

translational modifications so that they are

recognized by their binding partner. In addi-

tion, therapeutic proteins must be biologically

active and have the correct physiochemical

and immunochemical properties to elicit the

correct response from the patient.

To step back a bit, therapeutic proteins are

generally produced by cultured cells in large

bioreactors and then harvested and puri-

fied. The actual generation of the proteins

in the cell occurs along a complex pathway

that involves transcription of DNA in the

nucleus and translation of the protein in the

cytoplasm. The initial polypeptide chain is

further processed by folding and by post-

translational modifications to produce the

final biologically active protein. These pro-

cesses are very complex and, therefore, errors

can occur.

For this reason, during the production of

therapeutic protein, it’s vital to monitor all

aspects of the protein to ensure that its

sequence, structure, purity, and stability are

correct and consistent. Errors to the sequence

and the structure of the protein could result

in the drug not having the desired beneficial

effect in the patient, or even worse, these errors

could affect the safety of the drug and could

have the potential to cause detrimental effects

in the patient, such as eliciting an immuno-

Some might think it’s

sufficient to only characterize

the product during the

discovery phase and again at

the final product stage.

Product Characterization: A Primer NIBRT’s Jayne Telford provides an overview of biopharmaceutical analytics and their accompanying qualification and validation steps.

Page 44: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

42 BioPharm International www.biopharminternational.com November 2012

Boot Camp: Tech Guide

genic response, which could lead

to anaphylaxis—and that is obvi-

ously very dangerous. Basically, all

proteins need to be analyzed in

detail to ensure an active, pure,

stable, and safe drug that can be

administered to the patient.

One example of an error that

can occur in a therapeutic pro-

tein is the presence of nonhuman

glycosylation, such as alpha-gal

on the surface of the protein.

This non-human glycosylation

can be recognized by the body

as being foreign, and an immune

response can be elicited to get rid

of the drug. This incorrect prod-

uct would not be active in the

patient and could endanger the

health of the patient.

TIMING AND INSTRUMENTATIONBioPharm: When during the pro-

cess should such testing be per-

formed?

Telford: Some people might think

it’s sufficient to only characterize

the product during the discovery

phase and then again at the final

product stage; however, this is

not the case. It is advised by FDA

that the product be analyzed at all

stages during the processing path-

way, including during discovery,

upstream processing, downstream

processing, and right through to

the final product. This thorough

analysis is meant to ensure that the

product is consistent, active, stable,

safe, and pure from start to finish.

Analysis of all processing steps

also allows any changes to the

protein or other problems, such

as contamination, to be detected

immediate ly, thereby a l low-

ing for determination of where

the problem is occurring and

enabling faster investigations to

be performed. If analysis is only

carried out at one stage of a pro-

cess, it is difficult to pinpoint

the source of the problem and

lengthy analysis would be needed

to rectify the issue.

BioPharm: When performing

these analyses, what techniques

and instruments are most com-

monly used today?

Telford: There are many protein

characterization methods used

across the industry, all at dif-

ferent levels of complexity. The

simplest methods include pro-

tein estimation assays, sodium

dodecyl sulfate–polyacrylamide

gel electrophoresis (SDS–PAGE),

isoelectric focusing (IEF), and

Western blotting analyses. A pro-

tein estimation assay, such as

the Bradford assay, is a simple

method used to determine the

protein concentration of the sam-

ple. SDS-PAGE or IEF is frequently

used to separate the compo-

nents of the samples and to gain

information on the identity and

purity. Western blotting analysis

is routinely used to identify and

confirm the presence or absence

of the protein.

A not he r c o m mo n l y u s e d

method is capillary electropho-

resis. This method is useful for

ga in ing in format ion on the

size, charge, purity, and iden-

tity of proteins. In addition, this

method provides similar informa-

tion to SDS-PAGE and IEF, but it

tends to be easier to use, more

reliable, and more quantifiable.

There are other more complex

methods for protein characteriza-

tion, such as high-performance

liquid chromatography (HPLC)

and mass spectrometry. These

methods are extremely valuable.

Reversed phase chromatography,

size-exclusion chromatography,

and ion-exchange chromatog-

raphy are particularly useful for

separating the components in a

protein sample and for providing

information on the identity and

the purity of the protein.

Mass spectrometry analysis is

useful for confirming the amino

acid sequence of the protein and

for identifying the protein. There

are many other useful tools and

techniques that are widely avail-

able and widely used as well.

REGULATORY EXPECTATIONSBioPharm: There are many regula-

tions that the industry must fol-

low. Can you briefly discuss the

key FDA and EMA regulations

that describe requirements for

protein characterization of bio-

logics and what those organiza-

tions expect today?

Telford: Numerous regulations

exist for biologics in the United

States and European Union, and

they are categorized for different

types of therapeutic proteins and

for different types of production

pathways.

Some examples include guide-

lines for recombinant proteins,

guidelines for monoclonal anti-

bodies (mAbs), and more recently,

g u ide l i ne s fo r b io s i m i l a r s .

However, the guidelines for all

therapeutic proteins remain simi-

lar throughout the different cat-

egories and reference to the ICH

quality guidelines (that is, ICH

Q8 Pharmaceutical Development,

ICH Q9 Quality Risk Management,

and ICH Q10 Pharmaceut ical

Quality System) provide a good

basis for understanding what is

required (1–3).

To take mAb characterization

as an example, information must

be provided to regulatory author-

ities on the identity of the mAb

(i.e., sequence of amino acids,

structure and size, potency, het-

Glycosylation is the

most common post-

translational modifi-

cation of proteins.

Page 45: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

Integrated Database Reach

Print Magazine

Universe

eNewsletter

Universe

Both

CASTTM Audience Summary*

Print Magazine Universe...................................... 73,089

eNewsletter Universe .......................................... 92,679

Total Print Magazine and eNewsletter Universe ...... 114,333*

Pharmaceutical Science Conference Attendees...... 40,000

Pharmaceutical Science Webinar Attendees ........... 41,581

Total Pharmaceutical Science Conference and

Webinar Attendees .............................................. 72,988*

Total Unduplicated Print Magazine, eNewsletter,

Conference and Webinar .....................................180,313**

Conference

and Webinar

Attendees

Print and

eNewsletter

Subscribers

1.3 million

unduplicated

decision makers

Target Highly-Selective CASTTM Audience

CAST TM will help you create more effective

and innovative marketing campaigns that

zero in on the right markets with the right

customized products, including:

6�!�!3/(!00!./�6�!�++'/�6�3!��/%0!/

6���**!.���),�%#*/�6�,.+,.%!0�.4�.!/!�.�$�

6�2% !+/�6�3!���/0/�6�,+ ��/0/

All designed to achieve your specifi c

marketing objectives.

Find out how CASTTM can work for you by

contacting Mike Tracey at 732-346-3027

or [email protected]

www.biopharminternational.com

Want your marketing messages to reach the right

audience…every time?

*Publishers Data

**All duplicate records have been subtracted from these totals

Now you can drive demand and market

growth faster than ever before...

Meet CASTTM. The all new, highly targeted, data

driven, Custom Audience Segmentation Tool from

Advanstar’s Pharmaceutical Science Group.

CASTTM contains over 1.3 million unduplicated decision

makers from global pharmaceutical companies served

by our leading publications and conference brands.

With CASTTM we can help you…

6� �* !./0�* �0$!�/%5!��* �/�+,!�+"�4+1.�,+0!*0%�(�

target markets

6� �.!�0!�*!3�,�0$3�4/�0+�,+0!*0%�(��1/0+)!./�3%0$�

our customized products

6� �%.!�0�4+1.�)�.'!0%*#�)!//�#!/�0+�$%#$(4�0�.#!0! �

global audiences

6� �.%2!�-1�(%8�! �3!��/%0!�0.�"8���

6� �+�1/�4+1.�/�(!/�!""+.0/�+*�$%#$�-1�(%04�(!� /

Page 46: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

44 BioPharm International www.biopharminternational.com November 2012

Boot Camp: Tech Guide

erogeneity, glycosylation, immu-

nological properties, specificity,

cross-reactivity, whether there is

any aggregation of the product,

and process-related impurities).

Extensive analysis is required to

gather and provide this informa-

tion. Similar requirements are

outlined for other types of pro-

teins and processing types.

EMERGING METHODSBioPharm: Have you come across

any new or emerging analytical

techniques that interest you as

a training expert in bioanalyt-

ics? What gaps do you think still

exist in biopharmaceutical prod-

uct characterization?

Telford: Some emerging tech-

niques involve the analysis of

post- t ra nslat iona l mod i f ica -

tions of the therapeutic proteins,

which can be very d i f f icult.

Some examples of post-trans-

lational modifications are gly-

cosylat ion, phosphor ylat ion,

and oxidation, with glycosyl-

ation being the most common

post- t ra nslat iona l mod i f ica -

tion of proteins. It can be com-

plex and diverse, making the

analysis quite complicated, but

advanced glycosylat ion anal-

y s i s te ch nolog ie s i nc lud i ng

HPLC, ultra-HPLC, mass spec-

trometry, and capillary electro-

phoresis are emerging, thereby

enabl ing companies to ga in

increasing volumes of informa-

tion on glycosylation. However,

this a rea could benef it f rom

increased advances in technol-

ogy, especially in the analysis

of O -linked glycans, which are

much more difficult to analyze

than N-linked glycans.

Another area of interest is

protein aggregat ion analysis.

Proteins can aggregate under cer-

tain conditions to form dimers,

tr imers, and larger multimers.

These aggregations can affect

t he ac t iv it y, i m mu nogen ic-

ity, and stability of therapeutic

proteins. The detection of pro-

tein aggregates is complex, and

many techniques are emerging

to help analyze them, including

dynamic and multi-angle light

scattering, analytical ultracen-

trifugation, and size-exclusion

chromatography.

CHALLENGES AND BEST PRACTICESBioPharm: Validation of analyti-

cal methods is crucial to meet

regulatory requirements. Where

do qualification and validation

of methods come into play when

characterizing a product?

Telford: Qualification and vali-

dat ion are indeed important,

and challenging. All laboratory

equipment must be installed and

qualified (IQd) and operation-

ally qualif ied (OQd), and per-

formance qualified (PQd) prior

to validation of the actual test

methodologies. Val idat ion of

analytical methods is covered

well in ICH Q2 guideline on vali-

dating analytical procedures (4).

A significant part of valida-

tion involves ensuring that the

sof tware that is in use with

the equipment also meets the

requi rements of US Tit le 21

Code of Federal Regulations Part

11, entitled Electronic Records;

Electronic Signatures.

IQ, OQ, and PQ protocols and

reports need to be prepared and

approved for each piece of equip-

ment and for each test meth-

odology before analysis of the

product can begin. Once this has

been achieved, the test can then

be used to characterize the prod-

uct through the manufacturing

process.

BioPharm: What other chal-

lenges does the industry face in

this area?

Telford: One key cha l lenge

would be for companies to locate

skilled scientists as the advanced

met ho d o f p ro te i n c ha r ac-

terization can require special-

ized training for scientists to

be able to analyze the product

adequately and to allow them

to troubleshoot when problems

arise and to spot method-related

errors as they occur. Another

challenge is that the biologic

product characterization typi-

cally requires offline sampling,

which can take time and would

benefit greatly from advances in

online technologies.

Such advances could faci l i-

tate real-t ime analysis of the

product as it passes through

t he ma nu fac t u r i ng process .

For example, a sample could be

analyzed directly as it comes

out of a bioreactor. One other

major challenge, of course, is

the detect ion and prevention

of contaminat ion, a lthough

this would be helped by the

development of rapid analytical

methods for viral and microbial

contaminants at each stage of

the process.

REFERENCES 1. ICH, Q8 Pharmaceutical

Development (2009).

2. ICH, Q9 Quality Risk Management

(2005).

3. ICH, Q10 Pharmaceutical Quality

System (2008).

4. ICH, Q2 Validation of Analytical

Prodcedures: Text and Methodology

(1996). ◆

KEY TAKEAWAYS

t� 6OEFSTUBOE�UIF��

SFHVMBUPSZ�SFRVJSFNFOUT�

GPS�NFUIPE�WBMJEBUJPO�BOE�

FRVJQNFOU�RVBMJGJDBUJPO�

t� 1MBO�UP�UFTU�ZPVS�QSPEVDU��

GSPN�TUBSU�UP�GJOJTI�

t� 5SBJO�ZPVS�BOBMZUJDBM�TDJFOUJTUT��

Page 47: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 45

Boot Camp: Business Guide

Sve

ta D

em

ido

ff/G

ett

y I

ma

ge

s

Outsourcing stability storage is a highly

efficient way to cut costs, improve ser-

vices, and generate revenue by effective

cost management. Switching to an outsourced

stability storage facility could provide the benefit

of increased customer satisfaction and improved

efficiency. Outsourcing stability storage can free

up aspects of a company’s operations that might

not be running efficiently. Finding the right out-

sourcing partner, however, can be quite a chal-

lenge. Consider the following before arriving at an

off-site solution.

KEEPING STABILITY STORAGE IN-HOUSE IS COSTLYBy its nature, any controlled environment storage

facility needs a large investment in tailor-built

premises. Floor space comes at a premium and

much of the equipment needed has a large foot-

print, because it may have to be capable of accom-

modating bulk products. Typically, a resource

with several million cubic litres of environmen-

tally controlled storage space and storage suites

that can house thousands of cubic meters of prod-

uct is required.

Geographic location should also be considered.

The location of the facility should be well away

from natural disaster areas and be in a convenient

location for shipping options. Proximity to major

highways and airports can be a distinct advantage,

particularly considering the speed at which new

global markets are opening up.

Equipment investment

An average-sized storage room could

cost anywhere between $63,000 to

$190,000 to set up. A typical cabinet

could cost upwards of $23,500. With

separate rooms needed to suit differ-

ent environmental conditions, costs

can quickly mount up.

Walk-in stability room’s cabinets

and chambers should always be

available, with additional chambers

to be called upon for specific requirements such

as retained samples, secondary storage, quaran-

tine samples, freeze and thaw testing, and aero-

sol testing.

Design knowledge

Rooms and chambers need to be versatile. Through

their lifetime, these rooms will serve many pur-

poses including stability storage, commercial envi-

ronmental testing, corrosion testing, UV testing,

environmental stress screening, shock testing, tem-

perature and humidity testing, and photostability.

Ambient temperature and stability storage

should be available, regardless of the size of prod-

uct. Designing and building controlled-envi-

ronment rooms and chambers is a skill that is

developed over many years. Unique airflow tech-

nology systems should be incorporated to deliver

definitive environmental conditions.

REGULATORY CONTROLS Any facility working to cGMP guidelines should

incorporate chambers and rooms that are built

using the most modern techniques. They must be

mapped and validated for pharmaceutical stability

storage and biotech storage. Outsourcing compa-

nies can tailor these temperature and humidity

controlled rooms and chambers to suit individual

customer requirements as well as shelf life stud-

ies, intermediate testing, and accelerated testing

per International Conference on Harmonization’s

(ICH) Q1A (R2) guideline (1).

The assessment of drug substance stability is

a vital and essential aspect of the development

of pharmaceutical products. Stability testing is

capable of providing information on how envi-

ronmental factors such as temperature, humidity,

and light affect their quality over a period of time.

Data derived from a stability study enables recom-

mended storage conditions, re-test intervals, and

shelf lives to be evaluated and established.

It is the norm for controlled environment rooms

to be built and validated to provide climatic con-

Stability in Biopharmaceutical Storage Key considerations for choosing an outsourced sample storage facility.

Patrick Jackson is business

development director

at Vindon Scientific Limited.

Page 48: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

46 BioPharm International www.biopharminternational.com November 2012

Boot Camp: Business Guide

ditions specified in ICH guidelines.

This should include equipment to

allow for simulating conditions in

all four climatic zones for long-term,

intermediate, and accelerated testing.

An outsourcing facility should have

a comprehensive range of conditions

including ICH Photostability Option

1 and Option 2 studies, as well as

the capacity for customization. All

staff should be fully accountable and

provide a service that complies with

regulatory requirements.

TAILORING OF SERVICESAn independent storage facility

should be able to offer unique con-

ditions, as there are many instances

where drugs and drug-related prod-

ucts need to be stressed and tested

in situations outside ICH guidelines.

Any independent storage facility

should offer conditions specifically

suited to a company’s product. It

should meet the needs of companies

that require off-site stability storage,

redundant stability storage, short-

term growth needs, back-up storage,

or long-term stability storage.

Biopharmaceuticals, for instance,

are particularly sensitive to environ-

mental factors, making strict storage

conditions necessary for the main-

tenance of biological activity and

product integrity. The choice of tem-

perature for the storage of biological,

medical, and pharmaceutical materi-

als is dependent upon the sample

to be stored. An outsourcing facility

should offer the option for storing

materials under controlled ICH and

non-ICH conditions in a purpose-

designed storage suite. This service

will provide an extremely cost-effec-

tive solution for controlled-environ-

mental storage at temperatures that

should range from minus 196 ºC to

plus 55 ºC and relative humidities

from 15% to 90%.

Pure DNA will exist for long peri-

ods at 4 ºC in buffer solution. For

long-term storage, DNA is stored at

minus 40 ºC, while proteins are bet-

ter stored at minus 80 ºC and stem

cells are stored at minus 196 ºC. A

comprehensive cryobank will pro-

vide a solution for these and almost

all other storage applications.

CRYOGENICS There is an ever-increasing need

for cutting edge cryogenics in

today’s controlled storage world.

Cryopreservation is a process where

cell (biology) or whole biological

tissues are preserved by cooling to

low sub-zero temperature, such as

(typically) 77K or minus 196 °C (the

boiling point of liquid nitrogen).

At these low temperatures, any bio-

logical activity, including the bio-

chemical reactions that would lead

to cell death, is effectively stopped.

However, when vitrification solu-

tions are not used, the cell (biology)

being preserved is often damaged

due to freezing during the approach

to low temperatures or warming to

room temperature. These factors

are further proof that it makes eco-

nomic sense for pharmaceutical and

related industries to adopt an out-

sourcing partnership for their com-

plex cryobiological programs.

An outsourced controlled environ-

ment storage facility needs to provide

a full range of services to store and

manage biological materials at any

given temperature. From bar-coding

frozen vials through to relocating an

entire repository, the quality policy

should be reinforced by a positive

commitment. This starts with an

assurance that samples are stored in

a high security facility with conve-

nient access.

SAFE STORAGEThe storage of cells at temperatures

below minus 150 ºC is necessary to

preserve materials unaltered. This is

the point at which biological activity

ceases. Although mechanical devices

can be used to maintain tempera-

tures below this level, they can be

noisy and generate heat. They also

require liquid nitrogen backup to

avert disaster should the electricity

supply be disrupted or the freezer

fail. Liquid nitrogen is the most

logical choice for storage at tempera-

tures below minus 130 ºC. But there

are risks associated with its use. To

ensure there can be no cross-contam-

ination with the stored samples, it is

generally recommended that materi-

als are stored in the vapour phase

above the actual nitrogen, where

temperatures are in the region of

minus 150 ºC down to minus 178 ºC.

An efficient cryobank should

provide a management solution

for short-term, medium-term, and

long-term temperature controlled

storage. It should incorporate secure

storage and real-time tracking of

the stored samples and all safety

issues should be in strict accor-

dance with the latest regulations.

It is also essential that sites have

the staff and systems to offer fool-

proof biological storage conditions.

A purpose-built state-of-the-art bio-

repository should accommodate

biological samples, culture collec-

tions, microbial, viral seed stocks,

DNA and bone marrow, as well as

all-important stem cells and cord-

stem tissue.

Stem-cell and cord-blood storage

Stem-cell research is a market that

is in its infancy, but there is little

doubt that frozen sperm, eggs,

embryos, and stem cells being

stored at clinics and hospitals will

eventually become the norm across

the world. A good storage company

should already be constructing

cryobanks to satisfy the needs of

this emerging market. In the United

Kingdom, cord blood has become

the most frequent source of stem

cells for transplantation in chil-

dren. It is estimated that by the year

2015, there will be up to 10,000

cord- blood transplants worldwide

per year, using banked cord blood.

A vital aspect of this program will

be to develop repositories for the

storage of cells to guarantee contin-

ued successes in this exciting arena.

Page 49: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 47

Boot Camp: Business Guide

Long-term storage

There are some finely tuned meth-

ods of preserving biological mate-

rials by freezing and storing them

at ultra-low temperatures. The pro-

vision of long-term preservation of

biologics, reagents and specimens is

often required, together with a secure

facility for samples and laboratory

research materials. Ideally this should

cater for anything from a single box

of samples, up to virtually any vol-

ume, for the duration of any research

project and be capable of storing mil-

lions of samples, vials or tube racks.

VALIDATION A comprehensive storage facility

should have full regulatory compli-

ance to guarantee the integrity of

the samples it stores. They should

have the latest equipment to ensure

that the facility is technically fit for

purpose, meets the highest stan-

dards of environmental control,

and includes built-in redundancy

and emergency backup. Every piece

of equipment should also be pro-

tected by an individual preventative

maintenance schedule.

Constant monitoring is normally

performed with a paperless data

logging system that, ideally, should

also be compliant with the require-

ments of the US Code of Federal

Regulations. An ultra-low tempera-

ture storage facility of minus 70 ºC

to minus 85 ºC can be tailored to

the needs of biotechnology, phar-

maceutical, and agrochemicals

companies, including veterinary

industries and academic institutes.

Expertise in sample management

underpins the secure storage of sam-

ples and should include constant

monitoring of the storage conditions,

the ability to track and retrieve sam-

ples to GLP standards, and full trace-

ability in regulated environments.

Validation must be carried out

regularly to ensure that an environ-

mental room, cabinet, or cryogenic

freezer is capable of accurate and

repeatable performance. Temperature

and humidity levels should be moni-

tored at all times to incorporate an

identifiable audit trail.

SAMPLE TRACKING Managing hundreds of thousands

of samples for different companies,

with dissimilar protocols, requires

an efficient sample tracking system

to ensure that they are all handled

properly. When samples are out-

sourced properly, identification of

each sample is needed to prevent

confusion when removing the sam-

ples from chambers. Samples should

be clearly labeled, typically with

quantity, storage conditions, prod-

uct name, product code, lot number,

and date of manufacture. Reliable

software can also make a significant

difference when tracking pull dates,

sample locations, and quantities.

A comprehensive information

management system is at the heart

of all storage solutions. The inventory

management solution should enable

the user to record all data associated

with the samples including location,

temperature, and humidity, as well

as recording all user-defined informa-

tion. Every sample handled should

ideally be given a unique barcode

label and be recorded on the system.

This system significantly simplifies

handling, tracking, and processing.

Supplementary information relat-

ing to individual samples in terms

of movement within the facility,

time, and duration of storage vessel

openings and who accessed the ves-

sel must also be available within the

audit trail.

DISASTER RECOVERY PLANThe outsourced storage facility

should provide secure secondary stor-

age as an essential insurance policy.

A third party should retain duplicate

samples in a different but secure sec-

ond geographical location. This step

can help to eliminate the risk of a

disaster scenario destroying valuable

stability storage samples and causing

commercial damage.

COST CONSIDERATIONSAll this expertise can come at an

affordable price, usually levied as

a standard monthly or annual fee,

but charges for set-up, transac-

tions, or removing samples should

not be extra. The controlled-envi-

ronment facility should offer a

simple cost-effective arrangement

for a precise and intricately man-

aged service.

CONCLUSIONSControlled-environment storage is

a demanding business. It presents

many challenges including high

costs, management expertise, and

regulatory procedures. The decision

to outsource will depend on a com-

pany’s current resources and future

needs. Outsourcing provides quali-

fied and knowledgeable staff that

will keep abreast of topical develop-

ments, offering unique conditions

with state-of-the-art facilities to

match. Outsourcing is the logical

choice to navigate through the maze

of equipment, technologies, con-

trols, and procedures that lay ahead.

Choose carefully, and a strategy

can be developed that is not only

tailored to a specific organization,

creates and maintains a successful

and trustworthy relationship for

every program.

REFERENCE1. ICH, Q1A (R2), Stability Testing of New

Drug Substances and Products (Aug.

2003). ◆

KEY TAKEAWAYS

t� 0VUTPVSDJOH�QSPWJEFT�RVBMJGJFE�BOE�LOPXMFEHFBCMF�TUBGG�

t� "�DPNQSFIFOTJWF�TUPSBHF�GBDJMJUZ�TIPVME�IBWF�GVMM�SFHVMBUPSZ�DPNQMJBODF�

t� 5IF�TUBCJMJUZ�TUPSBHF�GBDJMJUZ�TIPVME�QSPWJEF�TFDVSF�TFDPOEBSZ�TUPSBHF�

Page 50: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

48 BioPharm International www.biopharminternational.com November 2012

Bioanalytical Best Practices

Imag

e: P

AS

IEK

A/S

cie

nce P

hoto

Lib

rary

/Gett

y. F

igure

s a

re c

ourt

esy o

f auth

ors

.

Assay Development and Method Validation EssentialsA 10-step systematic approach to analytical method development and validation can improve the quality of drug development.

Fundamental to all aspects of drug develop-

ment and manufacturing are the analyti-

cal methods. Analytical methods require

development, validation, and controls just as all

other product and process development activi-

ties. Measurement of API characteristics, the fac-

tors that influence them, and key impurities are

at the heart of product development for efficacy

and safety. This article discusses a systematic

approach for analytical method development

and validation that was developed in line with

the International Conference of Harmonization

(ICH) Q2 (R1) Validation of Analytical Procedures:

Text and Methodology, Q8 (R2) Pharmaceutical

Development, and Q9 Quality Risk Management

(1-3).

Historically, insufficient attention has been

paid to assay development, how it impacts the

product, ongoing release testing, and product

control. Simple coefficient of variation (CV)

calculations for assay precision are a necessary

but insufficient measure of assay efficiency and

may be misleading because CV has no relation-

ship to product acceptance and release testing

limits.

Assays and measurement systems must be

viewed as a process. The measurement pro-

cess (see Figure 1) is made up of methods, soft-

ware, materials and chemistry, analysts, sample

preparation, environmental conditions, and

instrumentation/equipment. Quality risk man-

agement techniques and statistical data analysis

techniques should be used to examine the pro-

cess of measurement and identify factors that

may influence precision, accuracy, linearity,

signal to noise, limits of detection and quanti-

fication, or any other assay attributes to achieve

optimal assay results (see Figure 2).

TEN STEPS TO ANALYTICAL METHOD DEVELOPMENTBased on ICH guidelines and the

author’s experience, there are 10

steps to analytical development and method

validation.

Identify the purpose

The purpose of any analytical method should

be clear. Will it be used for release testing or for

product/process characterization? What are the

target product profile parameters (ICH Q8[R2])

and critical quality attributes (CQAs) that the

analytical method is associated with? Are there

any CQAs that have no clearly defined mea-

surement method? What impurities need to be

measured, and what is the risk of not measur-

ing them? Is the assay correlated with other

analytical methods? How orthogonal is each

assay compared to other assays used to evalu-

ate the product. How does the assay minimize

or influence risk during drug development and

manufacturing?

Select the method used

There are many analytical methods across the

industry, and the method used should have

appropriate selectivity and high validity. Valid

analytical methods measure the condition of

interest. It is possible to have good precision

with poor measurement validity. It is possible,

for example, to measure the quantity of a pro-

tein without knowing how active the protein is.

Measures of activity and measures of quantity

need to be accurately considered and balanced

against other objective measures of the product.

Identify the method steps

Lay out the flow used in the analytical method

by using Visio or a similar process mapping

software to visualize the sequence used in per-

forming the assay. This layout will be used for

development, documentation, risk assessment,

and training. All steps should be listed and

detailed regarding the flow and use of plates,

materials, and chemistry. Identify steps in the

process that may influence bias or precision.

Thomas Little, PhD, is president of Thomas A. Little Consulting in

Highland, UT. [email protected].

Page 51: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 49

Bioanalytical Best Practices

Determine product

specification limits

The specification limits used to

control the release should be deter-

mined for the analytical methods

used for release testing. Limits may

be set using historical data and

industry standards, based on sta-

tistical k sigma limits, and/or tole-

rence levels or transfer function.

Limits need to reflect the risk to

the patient, CQA assurance, and

control the flow of materials in the

production of the drug substance

and drug product.

Perform a risk assessment

The analytical method risk assess-

ment (see Figure 3) is used to iden-

tify areas and steps in the analytical

method that may influence preci-

sion, accuracy, linearity, selectivity,

and signal to noise. Specifically, the

risk question to be asked is, “Where

do we need characterization and

development for this assay?”

Failure mode effects analysis

(FMEA) and or other risk assess-

ment methods may be used when

performing a risk assessment. In

addition to the traditional FMEA

approach, severity, probability

and detectability, the influence on

CQA and uncertainty to the risk

ranking should be added. Specific

questions of what may influence

precision or what may influence

bias or accuracy need to be exam-

ined. Each step in the analytical

method should be looked at from

this point of view.

Characterize the method

The development/characteriza-

tion plan for the assay should be

defined based on the risk assess-

ment. Determination of sample

size and sampling method are key

considerations. Assay development

can be broken into three steps: sys-

tem design, parameter design, and

tolerance design. System design

involves ensuring that the right

chemistry, right materials, right

technology, and right equipment

are being used. Parameter design is

typically done by running design

of experiments (DOEs) and mak-

ing sure that the right parameters

are selected at their optimal design

set point. Characterization of the

design space for precision and accu-

racy is a key assay development

outcome. Finally, the allowable

variation for key steps in the assay

must be defined to assure a consis-

tent outcome. Partition of variation

(POV) analysis is recommended to

further breakdown precision vari-

ation into its influencing factors

(4). Plate variation, for example,

must be considered when devel-

oping analytical methods. Failure

to understand plate variation and

other sources of assay error will

directly mix into the total variation

and will be linearly added to prod-

uct variation and increase limits of

quantitation and detection, effec-

tively reducing the assasy range and

adding to out-of-specifications rates

for product acceptance testing.

Complete method

validation and transfer

The method validation require-

ments should be defined. There

are many measures (e.g., amount

of API, activity of API, and impuri-

ties) of measurement performance

Measurement Process

Method Software Material People

Quality Measurements

Sample Prep Environment Instrument

Figure 1: Measurement process elements.

Criteria Minimum ProductProfile

Target ProductProfile

Optimal ProductProfile

Product QualityAttribute Name

Test orMeasurement

Definition

AttributeTarget

AttributeUpperLimit

AttributeLowerLimit

Attribute No.

12345123451234512345

CQA Purpose

Product Design Requirements and Critical Quality AttributesTarget Product Profile

INDICATION

ADMINISTRATION

DOSING and DURATION ofADMINISTRATION

POSSIBLE SIDE EFFECTS

Figure 2: Target product profile (TPP), critical quality attributes (CQAs), and

associated analytical methods.

ICH Parameter

Analytical Method Process Step and or Process Changes Risk Analysis

USL

Safety

Identify

Purity/impurity

Potency

Stability

Yield

Unit OperationNumber

Unit OperationNumber

Description

Baseline (optional)Change

(optional)Difference(optional)

Unit OperationDelta (Δ)

Potential Risk,Influence orFailure Mode

Severity and/orInfluence (1,3,5,7,10)

Probability and/orUncertainty (1,3,5,7,10)

Detectability(optional)(1,3,5,7,10)

Risk Score(RPN)

Severity xProbability

Only

Risk Score(RPN)

Severity xProb x Detect

Target LSLCQA/assay name

(release)Assay/Test name forCharacterization Only

1 5 3 5 00000000

0000000

752223334

Figure 3: Analytical method risk management example.

Page 52: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

50 BioPharm International www.biopharminternational.com November 2012

Bioanalytical Best Practices

that may be used in method valida-

tion (see Figure 4). Make sure there is

a clear identification of the require-

ments for each method when orga-

nizing the validation plan. Figures 4,

5, and 6 are adapted from Q2 (R1) and

identify the requirements to com-

plete the method validation.

Representative drug substance

(DS) and drug product (DP) materials

should be in place during validation.

Representative materials and stan-

dards will assure the limits of detec-

tion and quantitation have been

correctly calculated and validated

and will perform well when measur-

ing and testing actual product.

All method validation tests

should be conducted using the

correct sample size and sam-

pling method as defined in the

method standard operating proce-

dure (SOP). Acceptable results for

method validation of all analyti-

cal methods should be achieved.

Make sure acceptance criteria have

been defined for each validation

method variable. Aspects of the

assay should be modified so that

it can pass the validation testing

criteria. Finally, it is necessary to

determine whether the analytical

method is fit for use and ready to

transfer to other internal organiza-

tions or to external CRO/CMOs.

This is determined by meeting all

acceptance criteria for precision,

bias, and linearity. Equivalence

tests are typically used for method

transfer.

Define the control strategy

A clear control strategy needs to

be put in place once the assay has

been developed and validated (5).

The following questions should

be asked, “What materials will be

used for control or reference mate-

rials?” “How do you know the

standards are stable?” “What will

be used for tracking and trending

the assay so the true assay/plate

variation is known over time?”

“What will be used to adjust/

correct the assay once drift is

detected?” “How will one set of

reference materials be transferred

to another?”

Train all analysts

Train all analysts using the vali-

dated analytical method. If there

are concerns that the analyst may

have an effect on the results of

the analytical method, each ana-

lyst should run qualification tests

using known reference standards to

qualify and certify the analyst on

the method. Analyst method errors

may include sample selection, sam-

ple prep, weighing, mising, dilut-

ing, concentrating, location of

peak, injection method, and time

method.

Impact of the analytical method

Total variation can be expressed in

the following equation:

Standard Deviation Total = SQRT

(Product Variance + Assay Variance)

Method Validation List

Specificity

Repeatability (Intra Assay)

Intermediate Precision (Inter)Reproducibility (Inter Lab)

Visual

S/NLOD, LOQ (RMSE*K)/Slope

Linearity

RangeAccuracy

Precision

Detection andQuantitation Limits

RobustnessSystem Sutability

Method Transfer EquivalenceATP OOS Impact

Figure 4: Method validation list.

Definition

Assay Characterization Specificity Linearity Range AccuracyStandard: VALIDATION OF ANALYTICAL PROCEDURES Q2 R1, Nov 2005

Understanding of thefactors that influence themean and standarddeviation/CV of the assay.

To provide an exactresult which allows anaccurate statement onthe content or potency ofthe analyte in a sample.

The linearity of ananalytical procedure is itsability (within a givenrange) to obtain testresults which are directlyproportional to theconcentration (amount) ofanalyte in the sample.

The accuracy of an analyticalprocedure expresses thecloseness of agreementbetween the value which isaccepted either as aconventional true value or anaccepted reference value andthe value found.

For the establishment oflinearity, a minimum of 5concentrations isrecommended. Otherapproaches should bejustified. ICH Topic Q2(R1) Part II. Examinationof residuals will indicatewhere the linear range hasbeen established.

Minimum of 9 determinationsover a minimum of 3concentration levels coveringthe specified range (e.g. 3concentrations and 3 replicateseach of the total analyticalprocedure). ICH Topic Q2 (R1)Part II. 10 + determinations iseven better for accuracy.

The range of an analyticalprocedure is the intervalbetween the upper and lowerconcentration (amounts) ofanalyte in the sample(including theseconcentrations) for which ithas been demonstrated thatthe analytical procedure hasa suitable level of precision,accuracy and linearity.

Excipients, Concentrations,Assay Methods (# Dilutions).

Sample prep method,controlled impurities orsample matrix.

3-5 concentrations aretypical with 3 min.

Well characterized standardswith known potency etc.

QRM, Process Mappingand FR Matrix to identifykey factors in theanalytical method.

DOE, Full FactorialCustom Designs.

Assay or analyticalmethod designed todetect the specific drugattribute.

Fit Model and or Fit Yby X.

Linear fit, Ad Rsquare,equation (slope/intercept)and residuals plots.

Fit Y by X or Fit Model,Residuals.

Make sure concentrationsexceed drug applicationranges and refer to linearitystudy for range.

Fit Y by X.

Measure mean shift fromreference standard.

Fit Y by X, Distribution andGraph Builder.

Concentration.Typical Factors

Tip

JMP Platform

Recommended Dataand Analysis Procedure

Method Validation Quick Reference Guide

Figure 5: Method validation quick reference guide.

Page 53: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 51

Bioanalytical Best Practices

As the assay error r ises, the

total standard deviat ion also

rises. Using the accuracy to pre-

cision (ATP) model (see Figure

7), it is possible to visualize the

relat ionship of precision and

accuracy on product acceptance

rates. The ATP model shows how

changes in precision and accu-

racy impact product acceptance

rates and the assay error design

space. CV calculation is a good

measure of assay error; however,

it is not scaled to the acceptance

limits, it is scaled to the mean.

Rescaling the variation to the

release limits helps to clarify if

the variation in the assay is fit

for use. The number 5.15 is used

in the equation to represent 99%

of the assay error. Generally, a

percent of tolerance of less than

20% is considered an acceptable

result; more than 20% will result

in a high level of out-of-specifi-

cation release failures and should

be considered for further devel-

opment:

% Tolerance Measurement Error=

(Standard Deviation Measurement

Error*5.15)/(USL-LSL)

where USL is upper specifica-

tion limit and LSL is lower speci-

fication limit. The attention paid

to method development, valida-

tion, and control will improve

the quality of drug development,

patient safety, and predictable,

consistent outcomes.

REFERENCES 1. ICH Q2(R1) Validation of Analytical

Procedures: Text and Methodology,

(2005).

2. ICH, Q8(R2) Pharmaceutical

Development (2009).

3. ICH Q9 Quality Risk Management

(2006).

4. Little, T.A. Engineering Statistics and

Data Analysis-BPM (2012).

5. FDA, PAT—A Framework for Innovative

Pharmaceutical Development,

Manufacturing, and Quality Assurance

(2004). ◆

Accuracy to Precision

Accuracy to Precision

Prediction Profiler100.00%

% M

easu

rem

en

tW

ith

in T

ole

ran

ce

98.95%

0.704

Bias

Bias 01

1.5

1

-3 -2 -1 0 1 2 3

0.5

0.95 0.9973002 0.95

Precision

% Measurements Within Tolerance

% Measurements Within Tolerance

% Measurements Within Tolerance

Precision

Pre

cisi

on

Bias

Bias

Horiz Vert Factor

ContourResponse Current Y Lo Limit Hi Limit

Current X

Precision

0.994

90.00%

80.00%

70.00%

60.00%

50.00%

-3 -2 -1 0 1 2 3

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

Figure 7: Example of accuracy to precision modeling.

Type of analyticalprocedure

characteristics

Accuracy

Precision

Repeatability

Interm.Precision

Specificity (2)

Detection Limit

Quantitation Limit

Linearity

Range

-

-

-

-

-

- signifies that this characteristic is not normally evaluated+ signifies that this characteristic is normally evaluated

(1) in cases where reproducibility (see glossary) has been performed, intermediate precision is not needed(2) lack of specificity of one analytical procedure could be compensated by other supporting analytical procedures(3) may be needed in some cases

-

-

+

+

+

+ (1)

- (3)

+

+

+

+

+

+

+ (1)

-

-

+

+

+

-

-

-

+

-

-

-

+

IdentificationTesting forimpurities

quantitative limit

Assay- dissolution

(measurement only)- content/potency

Figure 6: What is required for method validation and when to use it (Ref. 1).

Page 54: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

52 BioPharm International www.biopharminternational.com November 2012

Manufacturing Best Practices

Ico

n I

ma

ge: R

efin

e T

ech

no

log

y

Simon Chalk is director of the

BioPhorum Operations Group,

[email protected]

The Transformation to Process-Centered Organization Is process-centered organization in biopharmaceutical manufacturing a stepping stone or a stumbling block?

The scientific and engineering chal-

lenges of producing biopharmaceuti-

cal products on an industrial scale,

historically, have required deep subject mat-

ter expertise to be deployed efficiently and

with direct responsibility for the result. This

modus operandi has remained intact because

most facilities were single product (i.e., high

volume, low variety) and underwent little

change. To compete in the current market-

place, however, pharmaceutical facil it ies

need to be increasingly multiproduct, flex-

ible, fast moving, and more customer centric

(i.e., low volume, high variety). The tradi-

tional command-and-control model is often

not responsive or agile enough to meet this

challenge.

Process-centered organization (PCO) struc-

tures have come into fashion over the past

few years. PCO structures realign organi-

zation building blocks with the value-add-

ing processes in the business. Instead of

functional hierarchies being the dominant

structure, people are organized into multi-

disciplinary teams whose goals are focused

on managing the end-to-end activities that

deliver value to their customer. Team size can

extend to many tens of members.

This concept has become best practice in

many industries where lean, fast

material and information flows

are crucial for survival. Removal

of c ross -funct ional boundar-

ies and an element of self-man-

agement and empowerment are

fundamental. When it works,

the value stream can be run with

fewer layers of management; head-

count efficiencies are increased;

and decision-making can be del-

egated more effect ively. Most

importantly, communication routes can be

short-circuited and the black holes found at

organization interfaces are often eliminated.

EXAMPLES OF PROCESS-CENTERED ORGANIZATION Several pharmaceutical companies have suc-

cessfully made the transition from a tra-

ditional organization structure to a new

progressive model. The result, in some cases,

has been the difference between survival and

eventual plant closure. In other cases, the

change has been a failure, and a reversal in

direction has ultimately been necessary.

In one g roundbreak ing example, an

inbound materials logistics process team was

formed out of separate functions from mate-

rials scheduling, warehousing operations,

quality assurance, and quality control. The

teams were co-located in an office adjacent to

the inbound material flow and given author-

ity to work together to meet common objec-

tives. A period of value stream mapping,

analysis, and improvement led to redesign of

the way of working, adjustments to the allo-

cation of responsibilities, and cross training

to enhance the range of skills in the team.

Prior to the change, receipt to release could

take anywhere from 7 to more than 40 days.

Material safety stocks reflected the length

and variability of these cycle times. After

the change, cycle times were consistently

achieved in the one-to-three day range. The

The command-and-control

model is often not responsive

or agile enough.

Page 55: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 53

Manufacturing Best Practices

team eagerly took up the chal-

lenge of empowerment and a cul-

ture of “self-direction” emerged

from the scheme. Support was

given to ensure the interpersonal

and team-working skills were in

place for the individuals. The

project became the starting point

for a more demanding redesign

in manufacturing areas where a

bottom-up approach was taken to

transform the wider operational

organization. In this case, struc-

ture followed strategy in a way

that supported irreversible cul-

tural change.

Another equally informative

case study showed how the orga-

nization structure in a facility

was redesigned in a radical way

with less emphasis on improv-

ing performance, ways of work-

ing, enlarging breadth of skills,

and more emphasis on manag-

ers becoming process flow own-

ers within a self-directed team

arrangement. The lines on the

organization chart were the cen-

ter of attention. In due course,

the change was found to be too

radical with insufficient focus on

business performance improve-

ment. Structure didn’t follow

strategy, and the organization

was changed back to a classic

line structure. After a period of

ref lection, the lessons learned

led to adopting a more effective

approach.

A common stepping-stone to

PCO is to use more of the matrix-

type structure where discipline

teams are linked to production

process teams by single contact

points. These dotted lines build

commitment and ownership to

the goals of the process team

without weakening membership

of the functional team.

THE ROLE OF SENIOR MANAGEMENTWhen it comes to making a dif-

ference, senior managers actually

have few levers they can pull.

I f they want to make change

quickly, the options reduce fur-

ther. The one change that always

remains open is organizat ion

change. Many senior manag-

ers choose to pull this lever as a

first option, particularly if they

are under pressure to improve

per formance, reduce cost, or

simply to be seen to be doing

something. Nothing gets atten-

tion more than an organization

review that potentially disrupts

the status quo. The problem is

that organization change can be

tricky and may not always lead

to the desired result.

So should the new organiza-

tion models and theories being

talked about be taken on board?

How re levant a re they to a

closely regulated environment

like pharmaceutical manufactur-

ing, and, if adopted, how should

they be implemented?

The dilemma is that from a

cultural perspective, the phar-

maceutical industry is rooted

in the “command-and-control”

mindset where strong oversight,

clear policies, and meticulously

defined procedures drive behav-

ior. The requirement is on the

employee to learn how a task is

to be done and to consistently

execute that task again and again

(and not necessarily to ask ques-

tions why). Managers and super-

visors are there to def ine the

task, to ensure it is done right

every time by suitably trained

people, and to solve problems

when they occur. Compliance

is key. Experts such as engi-

neers, quality professionals, and

support staff are also (highly)

trained to focus on their own

specialist areas.

There is no doubt that changes

where production operators are

g iven more responsibi l ity for

simple maintenance routines,

quality control, and batch sched-

ul ing, for example, conf ront

existing paradigms. This reality

can be seen to be controversial.

However, the professional ism

and capability of the people that

oversee the industries manufac-

turing sites are more than ade-

quate to ensure these changes

evolve in a way that maintains

produc t qua l it y and pat ient

safety.

For senior managers looking to

make an impact, it often makes

sense to leave the organization

change lever to last position on

the list. It then becomes an effec-

t ive tool with which to make

business improvement and cul-

tural change permanent.

Understanding the difference

between success and failure lies

not in organization theory, even

though it is important to take

change management factors into

account. It l ies in a “back-to-

basics” philosophy where mate-

r ia l paths are simpli f ied and

shortened, knowledge f lows to

where it is needed, people are

given a wider variety of skills

training, and wasteful non-value

adding activity is eliminated. ◆

Manufacturing Best Practices

Your opinion matters.

Send your questions, suggestions,

and comments for this column to

Simon Chalk at

[email protected].

Nothing gets

attention more than

an organization review

that potentially disrupts

the status quo.

Page 56: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

54 BioPharm International www.biopharminternational.com November 2012

SpotlightSpotlight

PERISTALTIC PUMPS OFFER

CONTAMINATION-FREE

PUMPING CAPABILITIES

Watson-Marlow’s peristaltic pumps provide simplified validation, improved processing, and reduced cost, and offer contamination-free pumping. The company’s Profibus enabled peristaltic pump is optimized for process monitoring in validated cGMP production. The pumps feature single-use tubing technology and can be used for a variety of applications including bag filling, buffer and media preparation, and live cell transfer.

Watson-Marlow’s new Bp pump features Profibus networking to the well-known benefits of peristaltic pumping, making it easy and efficient for customers to integrate this seal-less, valveless pump technology into their Profibus control system. Compared with non-networked pumps, the Bp range features Profibus compatible units that reduce engineering, hardware and installation costs, as well as offer remote diagnostics to help minimize plant downtime and maintenance costs.

Watson-Marlow

tel. 800.282.8823www.watson-marlow.com

TUBE AND CAP SYSTEM

FOR HANDLING BIOLOGICALS

The SaniSure Cap2v8 system is an integrated cap and tube system that provides aseptic transfer, storage, and sampling applications. Its one-piece molded cap reduces contamination and product loss that traditionally is associated with barbed fittings.

The flexibility of this product line allows customers the ability to custom configure systems with unique tubing sizes, lengths, and other accessories. The standard Cap2v8 components use C-Flex tubing, and the caps are made of polypropylene (other materials are available).

The SaniSure Cap2v8 system is designed for single-use, meets USP Class VI standards, is lot-traceable, is packaged in a certified ISO-7 cleanroom, and provides quick turnaround lead times on small or large-volume lots. With the option to have these components made into complete assemblies and shipped presterilized or not presterilized, customers have the flexibility of implementing technology specific to their specific needs.

SaniSure

tel. 877.777.3005www.sanisure.com

CHIRAL MEDIA NOW

AVAILABLE IN BULK

FOR PREPARATIVE

CHROMATOGRAPHY

Phenomenex now offers Lux Cellulose-1 and -2, unique and complementary chiral stationary phases in 10-μm particle size for batch preparative chromatography. The media includes a high-resolution screening set with a wide range of selectivity.

The new 10-μm particles are available both in Axia-packed preparative columns, and in bulk, enabling customers to pack their own columns for the most cost-effective purification methods. Lux chiral phases Cellulose-1, -2, -3, and -4 are already offered in 20-μm bulk media for use in batch preparative and simulated moving bed chromatography.

Lux Cellulose-1 uses cellulose tris (3, 5-dimethylphenylcarbamate) as the chiral selector. Lux Cellulose-2 provides a second chiral selector, cellulose tris (3-chloro-4-methylphenylcarbamate), providing a chlorinated stationary phase with complementary selectivity to Lux Cellulose-1.

Phenomenex

tel. 310.212.0555www.phenomenex.com

Page 57: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 55

Regulatory Beat

New Technology Showcase

BIOMANUFACTURING CAPACITYXcellerex’s experienced team helps clients

implement its FlexFactory biomanufacturing

platform effectively. The platform is intended

to overcome the limitations of conventional

biomanufacturing strategies. In addition, the

company offers a whitepaper that explains

how its approach to biomanufacturing

addresses various considerations facing the biopharmaceutical

and vaccine-production industries. Xcellerex, tel. 866.XCELLEREX,

www.xcellerex.com

SINGLE-USE BIOREACTORSEMD Millipore’s Mobius CellReady 200-L

bioreactor integrates several features

that are intended to provide ease of use,

reliability, and operational flexibility.

The unit contains a working volume of

40–200 L, which allows it to function as

both a seed and production vessel, and its standard design is optimized for

the cultivation of mammalian cells in suspension. EMD Millipore,

tel. 800.548.7853, www.millipore.com

LABORATORY SERVICESEurofins Lancaster

Laboratories enables

pharmaceutical and biopharmaceutical companies to advance candidates from

development through commercialization, ensuring regulatory compliance,

cost effectiveness, and achievement of timelines. Global pharmaceutical and

biotech services include viral clearance and safety, microbiology, biochemistry,

cell-bank manufacturing, release testing, raw-materials testing, and

mycroplasma testing. Eurofins Lancaster Laboratories, tel. 717.656.2300,

www.lancasterlabspharm.com

DEPTH FILTRATION SYSTEMThe Zeta Plus Encapsulated System is a single-

use depth filtration system designed for

the bioprocessing industry where upstream

cell-culture clarification or downstream

impurity removal is required. It features an

ergonomically designed large holder

that can be pivoted between horizontal and

vertical position to allow convenient handling, minimal footprint,

and minimal fluid spills. 3M Purification, Inc., tel. 203.238.8860,

www.3Mlifesciences.com

COMPLETE CHARACTERIZATIONThe ACQUITY UPLC H-Class Bio System from Waters

is suitable for the analysis of complex biomolecules

that require multiple chromatographic modes

for complete characterization—reversed-phase,

ion-exchange, size-exclusion, or hydrophilic

interaction—without hardware modifications.

The ACQUITY UPLC H-Class Bio System delivers

high resolution and sensitivity available with

sub-2-μm ultraperformance liquid chromatography. Waters,

tel. 508.478.2000, www.waters.com

CHROMATOGRAPHY DETECTORThe ViscoStar viscometer from

Wyatt Technology is designed as an

on-line chromatography detector

for determining specific and intrinsic

viscosities. According to the company,

the product has a software-based pulse

dampening correction algorithm that virtually eliminates pump pulses.

The instrument reportedly provides temperature control from 4 °C to 60 °C.

Wyatt Technology, tel. 805.681.9009, www.wyatt.com

Page 58: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

Company Page #

3M Purification Inc 17

ATMI Life Sciences 29

Agilent Technologies 27

Bio-Rad Laboratories Covertip, 55

Catalent Pharma Solutions 60

Cygnus Technologies 19

DASGIP Information and Process Technology GmbH 21

Nova Biomedical 59

Parenteral Drug Association 13

Parker Hannifin 31

Roche Diagnostics GmbH 5

Sartorius Stedim N America Inc 9

WTG–World Trade Group 33

Wyatt Technology Corp 2

Industry Calendar Ad Index

For event details and a complete calendar list, visit

www.biopharminternational.com/events

DECEMBER

3–4: Early Drug

Development Summit 2012

Location: Berlin, Germanywww.eddsummit.com

3–4: PDA/FDA Vaccines Conference

Location: Bethesda, MD www.pda.org*Listen to a podcast preview at BioPharmInternational.com

4–5: FT Global Pharmaceutical &

Biotechnology Conference

Location: London, UK www.ft-live.com/pharmabio

4–7: PDA/EMA Joint Conference

Location: Lisbon, Portugal www.pda.org*Meet our scientific editor at the event.

7: New York Pharma Forum

23rd Annual General Assembly

Location: New York, NY www.nypharmaforum.org/generalassembly

JANUARY 2013

7–9: Biotech Showcase 2013

Location: San Francisco, CA www.ebdgroup.com/bts

21–25: PepTalk:

The Protein Science Week

Location: Palm Springs, CA www.CHI-PepTalk.com

FEBRUARY 2013

18–20: 4th Annual World Drug

Delivery & Formulation Summit

Location: Berlin, Germany www.ddfevent.com

MARCH 2013

11–14: DCAT Week 2013

Location: New York, NY www.dcat.org

LAB EQUIPMENT

CELL LINES

Airfl ow Diagnostic Tools

FROM

ELEMENTS OF BIOPHARMACEUTICAL

PRODUCTION SERIES, FOURTH EDITION

Led by Dr. Anurag Rathore.

For anyone involved in or

planning to start process

development, characterization

and/or validation activities.

$129.95

save on these and other educational resources at www.industrymatter.com | 800.598.6008

Newly

Updated

Rapid Development ofHIGH PERFORMANCECHO Based Cell Lines

w w w . s e l e x i s . c o m

Page 59: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

November 2012 www.biopharminternational.com BioPharm International 57

Final Word

output, thereby reducing headcount.

Consumers benefited as well because

the vaccine backlog plummeted to

virtually zero. Improvements in

operations enabled a $500-million

increase in the sales of vaccines.

Another example involved a bio-

technology company that was

looking to increase the number of

high-quality drugs brought to mar-

ket by, in part, better leveraging its

vendors and partners to do every-

day, non-oversight work. This orga-

nization was specifically interested

in leveraging applied behavioral sci-

ence to identify and reinforce the few

critical behaviors that key performers

and leaders needed to consistently do

to ensure the sustainability of their

results to date. This company was

successful in sustaining its targeted

results by integrating sustainability

behaviors into their daily work and

embedding them within their busi-

ness management system.

STEPS FOR LEADING CHANGEBelow are some leadership tips for

change agility in the “new normal.”

Communicate about the change.

When major changes occur, such as

a merger or an acquisition, or pro-

cess changes related to QbD, ques-

tions run through everyone’s minds.

Leaders need to lead from the start

by helping people understand what is

happening, why it is happening, and

what it means to them.

Address your personal reaction to the

change. Regardless of whether you

are the CEO, a quality assurance

professional, or a front-line techni-

cian, when change occurs, people

in general can experience a range of

reactions (e.g., active or passive resis-

tance, or being enthusiastic about the

change but unable to develop specific

action plans) (4). Ultimately, the goal

is to become change-resilient and

create the best possible outcome for

you, your employees, and your end

users—the patients. Leaders often

need to attend to their personal reac-

tions to a change before they are able

to help others.

Support your team through the

change. During times of change, lead-

ers must focus on the business while

also tending to the staff. Successfully

leading in the new environment

requires helping team members by

first gauging their reaction, then

matching your coaching strategy to

those reactions.

Focus on key priorities. Change can

naturally lead to rumors, gossip,

and other distractions among staff.

Effective leaders need a plan to keep

employees focused on current and

future priorities and to ensure that

staff members feel successful about

what they are accomplishing. By

focusing them on a few short-term

priorities within their control, man-

agement can provide structure in the

midst of uncertainty. Make a point to

catch staff members “doing it right”

so that the focus on priorities is a

motivating experience for all.

Retain key staff. Leaders know that

it takes more than compensation to

keep their best performers engaged

during times of change. With an

average industry turnover of 14%,

during times of stability, and the

industry’s current talent shortage,

losing focus of key players in times

of change is not an option (5). One-

on-one communication with their

immediate supervisor that is person-

alized and career oriented can offer a

strong positive influence on a high-

performer’s decision to stay.

Track and resolve issues. Change cre-

ates “noise” in any system or process.

Leaders can gain loyalty from their

teams by tracking and resolving (or

escalating) questions and concerns.

CONCLUSIONLeading through change involves

more than good project manage-

ment and encouragement. Leading

through change is about helping staff

perform in new ways quickly and

consistently to achieve new results,

and helping them to be ready for the

next change that will come more

quickly than they might expect.

* Results achieved by Continuous Learning

Group clients.

REFERENCES1. T.J. Galpin and M. Herndon, “Welcome to

the Big Leagues of Change Management,”

in The Complete Guide to Mergers and

Acquisitions: Process Tools to Support

M&A Integration at Every Level, T.J.

Galpin and M. Herndon, Eds. (Jossey-

Bass Publishers, San Francisco, 1st ed.,

1999).

2. Right Management, “Ready, Get Set...

Change! The Impact of Change on

Workforce Productivity and Engagement,”

in Leadership Insights, Right Management,

Ed. (Right Management Inc., Philadelphia,

2009).

3. M. Beer and N. Nohria, Harv. Bus. Rev.

(May–Jun. 2000) 133–141.

4. J.M. Smith, “When ‘It’ Happens! at Work: 5

Action Steps to Make Change Work for You

(ChangeMatters LLC, Morgantown, WV, 2nd

ed., 2009).

5. M.Duggan, DrugTopics 154 (8) 21 (2010). z FIG

UR

E 1

IS C

OU

RT

ES

Y O

F T

HE

AU

TH

OR

S

Continued from p. 58

Perf

orm

ance

Transition Period (Times of Intense Change) Going Forward

Time

Performance Dip

Poor-PerformingOrganizations Suffer

High-PerformingOrganizations Maintain & Grow

Figure 1: Illustration of the performance dip avoided by “change-agile” high-

performing organizations during times of intense change.

Page 60: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

58 BioPharm International www.biopharminternational.com November 2012

Final Word

How to Manage Effective Leadership when Change is the Only Constant

There is a “new normal” in the bio/pharma-

ceutical industry. It is undergoing trans-

formational change because of initiatives

such as quality by design (QbD), tighter regulatory

enforcement, increased supply chain complexity,

patent expiration, new drug development (e.g.,

biologics, generics), and changing trends in patient

practices (e.g., more personalized treatments, vir-

tual monitoring).

Being able to effectively lead an organization

through these transformations is a core compe-

tence because it creates a change-agile organization

that is prepared to do things differently to achieve

the most optimal result during and after a transi-

tion. Such leadership allows organizations to avoid

performance dips (see Figure 1) that may occur in

productivity or morale during times of change (1).

For example, research shows that organizations

that do not manage change well are four times

more likely to lose talent (2). Effective change man-

agement enables successful adoption of changes

with minimal turmoil and brings to light systemic

factors necessary for sustainability of results.

Leaders of biopharmaceutical companies today

face the daunting tasks of leading their organi-

zations through continuous and overlapping

changes, and developing change agility through-

out the organization in a world where 70–90%

of strategic change initiatives fail to achieve their

business objectives (3). At the heart of change agil-

ity is the ability to judiciously, practically, and posi-

tively shift behaviors towards new expected results.

The most effective organizations take a disci-

plined approach to changing behavior, leverag-

ing the principles of applied behavioral science,

which is based on 100-plus years of peer-reviewed

research and scientific theory. The goal is to con-

sistently and practically reinforce those behaviors

that enable us to achieve our goals and win in the

market faster than the competition.

The prize? Organizations that effectively use a

behavioral focus to lead during times of change

have experienced significant results including:

improvement in crucial preventive mainte-

nance performance; increase in the use of plant

equipment leading to high-value product yield;

improved on-time shipments; and increases in

employee engagement*.

Take the example of a biopharmaceutical com-

pany that had launched several new vaccines, with

revenues projected to double from the previous

year. The company was forecasting a 60% increase

in units produced and a strong upside potential.

However, it was struggling with a culture that was

complacent and resistant to change, as well as a

laundry list of other challenges as listed below:

t� "�OFX� MBCPS�BHSFFNFOU�XBT� JO�QMBDF �CVU�NBOZ�

packaging operators were unhappy with it

t� .BOZ�TVQFSWJTPST�XFSF�OFX �XJUI�WBSJFE�MFWFMT�PG�

people-management experience

t� ����PG�FNQMPZFFT�IBE� MFTT� UIBO�POF� ZFBS�PG�

packaging line experience

t� 2VBMJUZ�BTTVSBODF� SFQPSUT� TIPXFE� UPP�NBOZ�

deviations in product quality, purity, or sterility

t� 5IF� SJHIU�GJSTU�UJNF�FSSPS�GSFF�EBUB�FOUSZ�PWFSBMM�

rate was only at 12%.

The company launched several efforts to address

these challenges, including organizational rede-

signs, Lean Six Sigma methodology, and team-

based Kaizen events. These changes helped, but

not everyone followed the desired behaviors

because operators and supervisors were not exactly

sure how to act differently—improvements were

not sustained. The leadership team realized that

something deeper was needed—a culture change.

Coaches helped company leaders and super-

visors to pinpoint specific behaviors that were

crucial to achieving genuine business results. The

team also provided behavior coaching, including

personal action plan development and execution,

with feedback collection. Lastly, coaches helped

leaders apply positive and corrective consequences,

including feedback that reinforced desired behav-

ior and discouraged undesired behavior.

After one year, the company saw a cost avoid-

ance (i.e., cost reductions from shortenend cycle

time and rework) of $7.3 million and when capac-

ity improved, it was able to in-source a chemical

that had previously been outsourced earlier, result-

ing in $1.3 million savings. Through the capac-

ity increase, fewer days were worked with more

Tracy Thurkow, PhD, is partner, Karen Gorman, is senior partner, and Paula

Butte is senior partner, all with the

Continuous Learning Group, tthurkow@clg.

com, [email protected], [email protected].

M. F

ree

ma

n/P

ho

toL

ink

/Ge

tty I

ma

ge

s

Continued on p. 57

Page 61: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

Advanced Technology Automated Cell Density/Viability Analyzer

BioProfile® CDVTM

Measures Cell Density, Cell Viability, Cell Size

High Resolution Trypan Blue Exclusion Assay at the Cost of

Low Resolution Assays

Accurate, Reproducible Counts in Less than 2.5 Minutes

Wide Measurement Range,Up to 80 Million Cells per mL

Automated, Walk-Away Operation

www.novabiomedical.com

Page 62: HELLOalfresco.ubm-us.net/alfresco_images/pharma/2014/12/26/c1033a07-65... · fragment to flow-through the column, ... Janet Rose Rea Vice President, ... Ajaz S. Hussain VP, Biological

GPEx‰ cell line

solutions.

faster to

clinic.

FASTER PATH FROM GENE TO CLINIC WITH GPEx‰ TECHNOLOGYWe can take you from cDNA

to high expression master cell

bank candidate in 5 months.

© 2

011

Ca

tale

nt

Ph

arm

a S

olu

tio

ns.

All

rig

hts

re

serv

ed

.

INTEGRATION FROM START TO FINISHFrom cell line engineering to

process development, cGMP

manufacturing, fill/finish and

packaging, we ensure your

biologics development success.

PROVEN CELL LINE DEVELOPMENT PERFORMANCEWe’ve successfully expressed

over 250+ different proteins,

including proteins used

worldwide in commercial

therapeutics and clinical trials.

Discover more solutions with Catalent.

Call: +1 888 SOLUTION (765 8846)

Email: [email protected] Visit: www.catalent.comDEVELOPMENT DELIVERY SUPPLY

We give you more, faster. Our GPEx‰ technology platform for mammalian cell line engineering integrated

with process development and cGMP biomanufacturing service solutions enable rapid biologics

development to help you get your drug to market quicker. With talented experts in technical development,

biopharmaceutical manufacturing, regulatory consulting, quality systems and project management, we

are the catalyst for your success. Catalent. More products. Better treatments. Reliably supplied.™

biologics