December 2012
Transcription
December 2012
Volume 25 Number 12 BioPharm International BioPharm INTERNATIONAL December 2012 The Science & Business of Biopharmaceuticals DECEMBER 2012 www.biopharminternational.com Employment Survey I Glycan Analysis I JOB SECURITY IN A CHANGING BIOPHARMA ENVIRONMENT Container-Closures RESULTS FROM OUR ANNUAL EMPLOYMENT SURVEY Volume 25 PEER-REVIEWED: Number 12 t%&4*(/$0/4*%&3"5*0/4 '03$0/5"*/&3$-0463&4 t7"-*%"5*0/0'"/&8 4$"-&%08/.&.#3"/& "%403#&3%&7*$& BUSINESS: t#&3,&-&:4637&: 3&7&"-45)&*/%6453:4 )*()&453*4,$0/$&3/4 t/"7*("5*/(&.&3(*/( '03&*(/."3,&54 HOW TO: t13&1"3&4".1-&4'03 $)30."50(3"1): t."45&3(-:$"/"/"-:4*4 ® Applied Biosystems resDNASEQ kits ® +LJKVHQVLWLYLW\KRVWFHOODNA TXDQWLWDWLRQXVLQJSURYHQ UHDOWLPHT3&5WHFKQRORJ\ ǩ6SHFLȌFLW\IRUWDUJHWKRVWFHOO '1$QRFURVVUHDFWLYLW\ZLWK XQUHODWHG'1$ ǩ2SWLPL]HGVDPSOHSUHSDUDWLRQ IRUTXDQWLWDWLYHUHFRYHU\IURP FRPSOH[PDWULFHV ǩ)RU&+2E. coli9HUR0'&. Pichia pastorisRU16 6KRSQRZDW lifetechnologies.com/resdnaseq Gibco FunctionMAX™ TiterEnhancer ® $UHYROXWLRQDU\DGGLWLYHIRU &+2IHGEDWFKSURFHVVHV IT TAKES ONE ǩ3URYLGHVXSWRGRXEOHWLWHUOHYHOV PRO TO SERVE ǩ+LJKO\FRQFHQWUDWHGS+QHXWUDO ǩ0LQLPDORSWLPL]DWLRQUHTXLUHG ANOTHER Let’s move forward together <RXQHHGDSDUWQHUWKDWǢVDVIRFXVHGDV\RXDUHRQ\RXUUHVXOWV)URPJROGVWDQGDUGSURGXFWVWR H[SHULHQFHGXSVWUHDPDQGGRZQVWUHDPSURFHVVFRQVXOWDQWV/LIH7HFKQRORJLHVFRQWLQXDOO\OHDGVWKH LQGXVWU\LQRIIHULQJSURGXFWVIRUSUHPLXPUHVXOWVLPSURYHGVDIHW\DQGIDVWWRPDUNHWHIȌFLHQF\ ǩ$FFHOHUDWHWLPHOLQHVDQGPD[LPL]HSHUIRUPDQFHZLWKGibco®FHOOFXOWXUHSURGXFWV ǩ ,QFUHDVHELRVHSDUDWLRQHIȌFLHQF\DQGSURFHVV\LHOGZLWKPOROS®FKURPDWRJUDSK\SURGXFWV ǩ(QKDQFHSURGXFWLYLW\WKURXJKDFFXUDWHVHQVLWLYHGHWHFWLRQDQGLGHQWLȌFDWLRQRIFRQWDPLQDQWV ZLWKApplied Biosystems® SEQUDSLGPROHFXODUPHWKRGV )RUH[FOXVLYHSURGXFWLQIRUPDWLRQHQUROOLQRXU HDUO\DFFHVVSURJUDPDW lifetechnologies.com/bioproduction 0D[LPL]H\RXUSURGXFWLYLW\—JRWR lifetechnologies.com/functionmax For research use only. Not for use in diagnostic procedures. ©2012 Life Technologies Corporation. All rights reserved. The trademarks mentioned herein are the property of Life Technologies Corporation or their respective owners. CO27358 1112 Invitrogen™ Applied Biosystems® Gibco® Molecular Probes® Novex® TaqMan® Ambion® Ion Torrent™ World Class Support Exceptional support for your Eppendorf New Brunswick ULT Freezer (SSHQGRUI1HZ%UXQVZLFNIUHH]HUV SURYLGH\RXSHDFHRIPLQGZLWKRXU XQSDUDOOHOHGVHUYLFHDQGVXSSRUW :LWKRXU7&$LQGHSHQGHQWUHPRWH WHPSHUDWXUHPRQLWRULQJV\VWHP \RXDUHJXDUDQWHHGUHOLDEOHVDPSOH SURWHFWLRQ7KHVPDOOSRGSURYLGHV HDFKXVHUDVHFXUHORJLQWRPRQLWRU WKHLU8/7IUHH]HUDQ\ZKHUHDWDQ\WLPH >5HDOWLPHFRQWLQXRXVPRQLWRULQJ >$ODUPQRWLILFDWLRQVYLDHPDLO WH[WDQGSKRQH >9HUVDWLOHRSHQV\VWHP >(DV\DQGLQWXLWLYHVHWXS >1RDGGLWLRQDOVRIWZDUHRU FRPSXWHUUHTXLUHG ZZZHSSHQGRUIQDFRPs 130.A1.0107.A © 2012 Eppendorf AG. BioPharm I N T E R N AT I O N A L The Science & Business of Biopharmaceuticals EDITORIAL K. A. Ajit-Simh Howard L. Levine President, Shiba Associates President BioProcess Technology Consultants Editorial Director Angie Drakulich adrakulich@advanstar.com Managing Editor Susan Haigney shaigney@advanstar.com Scientific Editors Amy Ritter, PhD, aritter@advanstar.com and Adeline Siew, PhD, asiew@advanstar.com Community Managers Stephanie Sutton ssutton@advanstar.com and Chris Allen callen@advanstar.com Director of Content Peter Houston phouston@advanstar.com Art Director Dan Ward dward@media.advanstar.com Contributing Editors Jill Wechsler, Jim Miller, Eric Langer, Anurag Rathore, Jerold Martin, and Simon Chalk Correspondents Hellen Berger (Latin & South America, hellen. berger@terra.com.br), Jane Wan (Asia, wanjane@live.com.sg), Sean Milmo (Europe, smilmo@btconnect.com) Herb Lutz ADVERTISING EDITORIAL ADVISORY BOARD BioPharm 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. Rory Budihandojo Manager, Computer Validation Boehringer-Ingelheim Edward G. Calamai Senior Consulting Engineer Millipore Corporation Senior Consultant BioProcess Technology Consultants Publisher Mike Tracey mtracey@advanstar.com National Sales Manager Steve Hermer shermer@advanstar.com European Sales Manager Richard Hodson rhodson@advanstar.com Market Development, Classifieds, and Recruitment Tod McCloskey tmccloskey@advanstar.com Direct List Rentals Tamara Phillips tphillips@advanstar.com Reprints The YGS Group AdvanstarReprints@theYGSgroup.com, 800.290.5460 ext 100 or +1.717.505.9701 ext 100 Vice President Sales/Group Publisher Russ Pratt rpratt@advanstar.com Anurag Rathore PRODUCTION, MARKETING, CIRCULATION Biotech CMC Consultant Faculty Member, Indian Institute of Technology Production Manager Jesse Singer jsinger@media.advanstar.com Audience Development Manager Nidia Augustin naugustin@advanstar.com Managing Partner Pharmaceutical Manufacturing and Compliance Associates, LLC Hans-Peter Meyer John Carpenter K. John Morrow Professor, School of Pharmacy University of Colorado Health Sciences Center President, Newport Biotech 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 VP, Innovation for Future Technologies Lonza, Ltd. Barbara Potts Director of QC Biology, Genentech Tom Ransohoff 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 Denny Kraichely President, Statistical Outsourcing Services Associate Director Johnson & Johnson Gary Walsh 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. 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 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 ©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: mcannon@advanstar.com. 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. BioPharm I N T E R N AT I O N A L Contents Volume 25 Number 12 December 2012 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. PEER-REVIEWED FEATURES FEATURES MEMBRANE SCALE-DOWN A New Scale-Down Membrane Adsorber Device for Process Development and Validation SURVEYS Job Security in a Changing Biopharma Environment ON THE WEB www.biopharminternational.com Social Media Amy Ritter Nathalie Frau, Martin Leuthold, Amit Mehta, Kome (Kevin) Shomglin, and Rene Faber Results from the 2012 employment survey. 12 The development of an ultra scale-down anion exchange membrane adsorber. 18 Optimizing Global Biopharmaceutical Operations Through Risk Mitigation and Management Follow us on Twitter@BioPharmIntl CONTAINER CLOSURES Closures for Pharmaceutical Preparations: A Review of Design and Test Considerations Tim Sandle The author examines the use of closures for products intended for injection. 32 Phil Kaminsky, Jiyang Liu, and Julia Olsen-Claire A UC Berkeley survey provides insight into biopharma’s risk concerns and strategies. 38 GLOBAL MARKETS Navigating Emerging Markets Jill E. Sackman ANNIVERSARY RETROSPECTIVE A 25-Year Retrospective on Computer Validation An introduction to a new series on manufacturing within global markets. 46 Join our BioPharmInternational Group BioPharm Bulletin Subscribe to the one industry newsletter focused on the development and manufacturing of biotech drugs and vaccines. Catch up on regulatory actions, new technologies, industry deals & more. biopharminternational.com/subscribe 8 Cover: Atomic Imagery/Getty Images COLUMNS AND DEPARTMENTS 6 From the Editor Keeping tabs on crucial medicines should be part of emergency-preparedness plans. Angie Drakulich 7 Global News 10 Regulatory Beat White House and Congress likely to struggle over funding. Jill Wechsler 48 Boot Camp: Tech Guide NIBRT’s Pauline Rudd on what to expect when performing glycan analysis. 52 Bioanalytical Best Practices Preparation of biological samples for chromatographic analyses. Roger N. Hayes 55 Final Word Can postapproval FDA filings immunize pharma companies from patent lawsuits? Kevin Murphy and Andrew Nason 57 Ad Index/Marketplace BioPharm InternationalJTTFMFDUJWFMZBCTUSBDUFEPSJOEFYFEJOrBiological Sciences Database (Cambridge Scientific Abstracts)rBiotechnology and Bioengineering Database (Cambridge Scientific Abstracts)rBiotechnology Citation Index (ISI/Thomson Scientific)rChemical Abstracts (CAS) rŞScience Citation Index Expanded (ISI/Thomson Scientific)rWeb of Science (ISI/Thomson Scientific) 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. 4 BioPharm International www.biopharminternational.com December 2012 Beat your best score With enhanced Solubility thanks to a wide range of advanced products and technologies Converting a promising API to a final drug can be as tricky as hitting a hole-in-one, with poor solubility as the biggest challenge. EMD Millipore helps you to turn this challenge into a solution, e. g., with Meglumine, our API grade counterion in ICH/Q7 quality. Along with products for solid dispersions, we offer innovative technologies such as API modification and hydrogel formation to make your pipeline more efficient. Find out how to beat your best score at www.emdmillipore.com/bioavailability EMD Millipore is a division of Merck KGaA, Darmstadt, Germany From the Editor When Disaster Strikes at Home T Angie Drakulich is the editorial director of BioPharm International. Keeping tabs on crucial medicines should be part of consumers’ and manufacturers’ emergencypreparedness plans. 6 he editorial offices of BioPharm International are based in New Jersey, so when Hurricane Sandy hit last month, we were all affected. Flooding and high winds destroyed many coastline homes along with well-known destinations across Atlantic City, the Jersey Shore, and the neighboring Manhattan boroughs. Suburban communities and businesses (including many US biopharma manufacturing headquarters) across the state were without power for days and schools were shutdown for a week or more in several counties. President Obama declared the state, along with New York, a major disaster area. The entire ordeal was quite surreal, and our team considers ourselves to be lucky that we made it through unscathed. Our thoughts go out to those who are still recovering from the storm and trying to get back to normal. Many common questions arose during the hurricane and its aftermath, including how long food products could be considered safe to eat without refrigeration and where the closest open gas station was to refill generators. Pertaining to our industry were questions about medications that required refrigeration (primarily injectables and liquids), prescriptions that needed refilling (many doctors’ offices and drugstores were closed due to flooding and power outages), and drug products that had gotten wet or been lost in the storm. Fortunately, FDA has a consumer webpage devoted to drug safety after a natural disaster, whether it be exposure to fire, unsafe water (key for drugs that have to be reconstituted), or lack of refrigeration (www.fda.gov/Drugs/EmergencyPreparedness/ ucm085200.htm). Certain life-saving drugs (e.g., insulin) can be used even if not cold as long as they are not past their expiration date, says the agency webpage. There is also FDA information on medical devices and their safety during a crisis, and even an agency guidance aimed at sales representatives on how to deal with lost or stolen drug samples in the aftermath of a disaster (looting in general was quite prevalent after Hurricane Sandy). There are also some rules of thumb to follow in any emergency-preparedness plan. Many patient advocates recommend having on hand a 30-day supply of regularly used medicines (both prescription and over the counter), as well as extra quantities of devices needed to administer medications. But as a consumer, it seems there should be even more information available for those unfortunate times when something out of human control affects the medications we need. As an asthmatic, for example, I was concerned about my inhaler’s pending expiration date and not being able to get a refill without having to go the emergency room, which by the way, would have been impossible given that the end of my street had a 60-foot tree and power lines across it. Drug labels are already quite lengthy, but it may be worthwhile to add a few more details. For instance, language may focus on how long a cold-chain medicine can go without refrigeration, or how long a drug exposed to excessive heat can be considered safe, or the reasons behind drug-product expiration dates. Some of these details may be provided in medication guides that come with drug products, but even those of us in the pharma industry know that the majority of end users do not read those packets in full or keep them. This is probably an area where FDA and pharmacists can do a better job educating consumers about the importance of medication guides. Another piece of information worth including on labels may be where to find information during emergency situations. As so many discovered during Hurricane Sandy, we rely heavily on technology to get our information. Smart phones, in particular, became indisposable in the aftermath of the storm when power outages held hostage traditional television, phone, and Internet access. Pharma companies could consider, for example, having active Twitter feeds during emergencies so that people can still access crucial information. Various disasters and crises are bound to affect the global community in the future. Now is the time to think about the crucial information we will need to have on hand—and attached to our medications—when that next time strikes. z BioPharm International www.biopharminternational.com December 2012 Global News Understanding Cellular Reprogramming A publication in the Nov. 21, 2012 issue of Cell examines the mechanisms underlying the reprogramming of somatic to become induced pluripotent stem cells (IPSCs). IPSCs have engendered much excitement as potential tools for disease modeling or for regenerative medicine, but the methods used to produce them are inefficient and time consuming, limiting their commercial potential. The method pioneered by Shinya Yamanaka of Kyoto University, which earned him a Nobel Prize, involves adding four transcription factors, Oct4, Sox2, Klf4, and c-Myc, to a somatic cell, usually a skin cell (i.e., fibroblast). A team of scientists from the University of Pennsylvania looked at where on the chromatin those factors were bound 48 hours after transfection to understand the sequence of events that leads to reprogramming. They found that Oct4, Sox2, and Klf4 bound enhancer regions of the chromatin distant from the genes they regulate. The authors suggest that these transcription factors act to open closed chromatin structures, allowing transcription machinery to access the DNA. C-Myc appeared to act by enhancing the binding of the other factors to the chromatin. The researchers also found large regions of the genome where the transcription factors would not bind at 48 hours, but which were activated at a later time. The DNA-binding proteins, called histones, associated with the refractory regions were found to be chemically modified with a modification called H3K9me3. Moreover, blocking the enzyme that produced the H3K9me3 modification was found to accelerate the reprogramming process. By understanding cellular reprogramming at the genetic level, scientists will be able to better control the process. —Amy Ritter Source: A. Soufi, G. Donohue, and K. Zaret, Cell online, DOI:0.1016/ j.cell.2012.09.045, Nov.15, 2012. Report from South Korea Four domestic companies filed a suit against the Ministry of Health and Welfare of South Korea claiming that the recent price cuts made by the ministry have affected their businesses. On April 1, 2012, drug prices were reduced by an average 17% and affected the prices of 6506 drugs across the board. The first cut, announced in 2011, decreases the price of off-patented drugs 30%, with the price of the first generic drug set at 60% of the price of the off-patented drug. Originally, prices of off-patented drugs decrease by 20%, while the first generic version is set at 68% of the off-patented drug. The second cut reduces the price of drugs due to illegal rebates. The combination of the two cuts means that certain drugs will undergo double-price reduction and the final price rate can be up to 53.55%. Pharmaceutical companies are crying foul because these cuts would have a direct impact on their business profits even though the agency claimed that this move would ensure market sustainability and eradicate the problem of illicit rebates. In fact, a Sinhan Investment & Securities source has indicated that the majority of companies experienced a 20% fall in profits since the policy took effect. In response to their declining fortunes, some companies have opted to increase prices of their over-the-counter (OTC) drugs while others have boosted their R&D expenditure. Domestic companies such as Dong-A Pharmaceutical and LG Life Sciences have committed 22% and 19%, of net sales on R&D, respectively. Cher Boon Piang, an analyst for Asia Pacific Pharmaceutical and Healthcare of Business Monitor International (Asia), says, “Given the price cuts, companies may withdraw drugs that are not profitable. Local companies may even move away from generics. There is also a shift towards biosmiliars that opens opportunity for companies.” In October 2011, Dong-A Pharmaceutical joined hands with Tokyo-based Meiji Seika Pharma to build a biosimiliar plant in Songdo. Recently, local companies Yuhan Corp and Teregen ETEX are collaborating to commercialize the provision of individual genome services. The recent US-Korea free-trade agreement (FTA) has also crippled domestic players as it contains provisions protecting the intellectual property rights of original drug developers. For instance, the Korean agency has to inform original manufacturers if there are companies looking to produce generic versions. Companies are denied market approval if an objection is posed by original drug manufacturers and when the claimed patent exists. It is also mandatory for the generic manufacturer to provide safety and efficacy information to ensure that the generic version does not infringe on the original one. Perhaps a gradual price cut would have helped in alleviating pressures faced by industry players in South Korea. Cher says, “In general, companies face[d] both financial and time issues when two price cuts were introduced in 2011. If price reduction is gradual and made known to companies in a timeframe that prepares them for such reduction, these companies can draft and implement strategies to minimize the impact of price reduction. Moreover, it is easier to have short-term solutions against gradual price cuts compared to a larger one-off reduction.” December 2012 www.biopharminternational.com BioPharm International 7 Kevin Forest/Getty Images Discovery Pipeline Global News Cher points out that price cuts cannot be the only solution to contain rising government expenditures and it is also unfair to shift the burden onto companies. Instead, the government should look into ways to cut expenditure by subsidizing only the necessary and/or increase premiums. These strategies may create a negative impression, but they are essential if a country is not doing well economically. In addition, the burden of healthcare cost should rest on individuals instead, he adds. Clearly, the objectives of the government and the pharmaceutical industry players differ greatly. On one hand, the government seeks to lower healthcare costs. On the other hand, industry players are looking for ways to maximize profits. Asked if a balance can be struck between both parties, Cher says, “The level of compromise is dependent on the attractiveness of the market. Typically, an attractive market allows the government to push through its policies as companies are willing to forgo higher profit margins in exchange for sustainable growth over a time period.” The South Korean market is characterized by its aging population and an affluent population. Growth potential is limited as it has evolved to a developed market and industry players expect it to have established regulations. Therefore, it makes sense that industry players are pre-alerted of any policies in the government’s agenda. For example, industry players were informed in 2010 of the price disclosure policy to take effect in 2012. In Japan, industry players understand that it is the usual practice that price cuts occur once every two years. Despite its fragmented market and the pricecut setback, South Korea is ranked among the world’s top 12 with $8 billion annual sales. Cher adds, “In the long run, it has the necessary ingredients to continue with its successful pharmaceutical industry, strong support for innovation, the willingness of the private sector to explore these innovative technologies and the demographic profile also supports increased drug usage. The conflict between the government and industry will definitely arise again in the future, but we believe the two parties will reach a compromise. “ —Jane Wan is a freelance writer based in Singapore 8 BioPharm International A 25-Year Retrospective on Computer System Validation Throughout BioPharm International’s 25th anniversary year, we have looked back at articles published in the first volume of the journal. This month, Sharon Strause, an industry consultant, provides a look back at “Computer System Validation Part I: Testing and Verification of Applications Software” by Leonard J. Goren. Computer system validation has changed in the past 25 years as technology has become more complex. The majority of the documentation requirements and software tests Leonard Goren listed in his article, “Computer System Validation Part I: Testing and Verification of Applications Software,” are still applicable today. One of the key missing procedures required to execute computer system validation well is the supplier management process, which should include the requirements for a vendor audit. A vendor audit determines the vendor’s capability of producing a software application that can be validated for a regulatory company’s use. A vendor audit verifies how the code was developed, documented, and tested in all stages of the development process. A vendor audit determines the quality system procedures that a vendor has in place to ensure a well-developed software application. The audit determines how code is managed (i.e., configuration management), reviewed, internally documented, tested for the use and misuse of its software, and changed. The vendor audit captures how data produced by the software meet the requirements document, which starts the process of a vendor audit. CGMP requirements are established in the company’s requirements document for the software application including any audit trail functionality; how the code is managed from an infrastructure perspective (e.g., local, wide area, and web networks); and disaster recovery process (i.e., backup process and offsite code management). A thorough vendor audit showing good development practices will then allow a company to qualify the software’s installation on their equipment, and validate it for their use according to the computer system validation procedures in place at the company. If practices at the vendor are not quality capable, then a company can either chose another vendor or know in detail the additional requirements that would need to be completed to fulfill the requirements of computer system validation. This could mean thoroughly testing the software for accuracy of the stated requirements of the software as it is received before ever beginning the actual “validation for use” that is in place at the company. The other crucial document that must be in place prior to assessing any vendor is the requirements document. Goren addresses the functional requirements in his 1988 article, but today, a requirements document includes functional, technical, and regulatory requirements for a software application. A completed requirements document helps to determine the types of applications that should be assessed and also the types of testing that may be required to ensure workability of the software. It’s an important starting point in the computer system validation process. Having a computer system validation process that starts with requirements and includes a good process for supplier management and auditing can help to minimize the validation of an application or make it more complex. z —Our Retrospective series has included updates on separations technology, mammalian cell culture, industry perceptions, orphan drugs, mAbs, GMP training, cleanroom management, and more. For a complete list of Retrospectives and their original 1988 articles, visit BioPharmInternational.com/Retrospectives. www.biopharminternational.com December 2012 Choose Choice. Sustained-Release Immediate-Release Taste Masking Lyophilization Aseptic Filling Multi-therapy Solubilization No other partner gives you more formulation options – royalty free. Get access to the industry’s widest range of complex formulation technologies for small molecules and biologics, and benefit from expertise forged over thousands of projects. At Patheon, we’re not tied to any technology. That means science alone drives the development of an optimal formulation, and you’ll never pay us a royalty. Our commitment is to your success. Choose choice – choose Patheon. Visit www.patheon.com Call +1 866-PATHEON Email doingbusiness@patheon.com ©Patheon Inc. All rights reserved. Published 11/12 PATH0214R2 Regulatory Beat Research Policies Pose New Challenge for US Administration White House and Congress likely to struggle over funding for bio/pharmaceutical regulation. 10 Executives at bio/pharmaceutical companies are watching closely at how tax and budget proposals will affect corporate tax rates and investment. commit to these new programs in a period of political uncertainty. SPENDING CUTS AHEAD The dark cloud looming over all these programs is the year-end “fiscal cliff,” with nearly $500 billion in tax increases and spending cuts scheduled to begin Jan. 1, 2013 unless Congress acts. Executives at bio/pharmaceutical companies are watching closely at how tax and budget proposals will affect corporate tax rates and investment, as well as the specific funding for FDA, the National Institutes of Health (NIH), and other activities important to biomedical innovation and healthcare coverage. All sides acknowledge the crucial need to reduce both public and private outlays for US healthcare, and drug prices and reimbursement are a prime target, particularly related to outlays for federal government health programs and Medicare Part D. House Democrats have pressed for added rebates on drugs purchased by Medicare drug plans for low-income “dual eligible” seniors, which could total more than $100 million over 10 years. There also are budget proposals on the table to reduce federal spending on drugs for federal government employees, as well as other government health programs. BioPharm International www.biopharminternational.com December 2012 Digital Vision/Getty Images T he calm after the heated election battle this year has been brief due to pressure on policymakers to tackle overwhelming budget issues. With Republicans maintaining tight control over the House of Representat ives but losing g rou nd in the Senate, much depends on the abilit y of President Obama to engineer some kind of “fix” to the mounting deficit during the year-end “lame duck” Congressional session. Healthcare policy was a key point of dispute during the election campaign, marked by promises of better coverage and predictions of soaring costs by both candidates. Now, scheduled funding cuts and significant tax increases are expected to play a large role in shaping the reform program. The Obama v ictor y ended prospects of wholesale repeal of the Affordable Care Act (ACA). House Republicans will continue to challenge various requirements of the healthcare legislation, but key provisions for pharmaceutical companies, such as rebates on drugs for seniors in the Part D coverage gap and authorization for biosimilars, are unlikely to change. More broadly, the promised expansion of coverage to some 30 million prev iously uninsured A mer icans will move forward, although with consumers paying higher premiums and cost-sharing to cover ever-rising healthcare costs. That sets the stage for significant growth in the market for brand-name drugs. The Department of Health and Human Jill Wechsler is BioPharm Services (HHS) is working hard to International’s Washington editor, meet a host of deadlines and timeChevy Chase, MD, 301.656.4634, frames for establishing exchanges, jwechsler@advanstar.com. defining benefits, and expanding Read Jill’s blogs at Medicaid, much of that involving PharmTech.com/wechsler. states that have been reluctant to Regulatory Beat The Obama victory offers some stability for FDA, as the agency continues to implement the FDA S a fet y & I n novat ion Ac t a nd struggles to find a middle ground between speeding untried new medicines to patients and protecting the public from undue risk and harm. Although there won’t be a wholesale change in executive branch leadership, many top administration officials are likely to move on to other roles, and extensive cuts in the 2013 budget could undermine many FDA projects. An 8.2% cut in the FDA budget, as proposed under the sequester process, would reduce FDA’s 2013 budget by $320 million and prompt the agency to lay off approximately 1000 employe e s, accord i n g to con s u lt a nt Steven Grossman, publisher of FDA Matters. Even without such a severe, across-the-board cut, which could jeopardize FDA’s ability to collect user fees from pharmaceutical and medical device ma ke r s, t he F DA budget w i l l remain vulnerable to pressures to reduce federal spending for some years to come. Severe reductions in NIH funding, moreover, would jeopardize the pace of new drug and biotech discovery and support for clinical research that is key to spurring innovation needed to fill the depleted new drug pipeline. The biomedical research community is highlighting the importance of both FDA and NIH in protecting public health for patients at home and around the world. CONGRESS TACKLES COMPOUNDING Meanwhile, mounting deaths from contaminated steroid injectables made by a Massachusetts compounding pharmacy are focusing attention on the need for broader FDA legal authority in this and other areas. The need to reauthorize animal-drug user fees in 2013 is expected to provide a vehicle for legislation that would better secure the prescription drug supply chain and also address drug compounding oversight. (For more information on this subject, view “Compounding and FDA Regulation” on w w w. BioPharmInternational.com.) The ongoing fungal meningitis outbreak had sickened more than 425 individuals and caused over 30 deaths, as of early November. Rep. Edward Markey (D-MA) has proposed legislation to enhance FDA oversight of compounding pharmacies, and lead House and Senate committees held hearings right after election day to address the response by FDA and state regulators and to analyze actions by the offending compounder, the New England Compounding Center (NECC). Markey’s bill clarifies FDA’s right to inspect and regulate large compounders that qualify as drug manufacturers. Small compounding pharmacies would continue to operate under state licensing, and FDA could issue waivers to operators responding to drug shortages and public health crises. Compounded drugs have to be labeled that they have not been tested for FDA safety and efficacy standards, and FDA has to publish a list of unsafe or ineffective drugs not suitable for compounding. FDA regulation of compounders has been a thorny issue for decades, as previous efforts by the agency to impose stricter rules on compounders have been struck down by the courts. But the recent crisis has reopened the debate over the adequacy of state versus federal regulation of pharmacies and when compounding qualifies as drug manufacturing. Efforts by FDA and Massachuset ts reg u lators to shut dow n N ECC and its sister company, Ameridose, highlight the links between drug shortages and compound ing. FDA Commissioner Margaret Hamburg noted that the agency is working hard to minimize shortages in important Ameridose products used in surgery and to prevent congestive heart failure. Yet, former FDA official Scott Gottlieb also commented that too-tight FDA regulations have led to shortages of low-cost injectable drugs, prompting hospitals and patients to seek a lte r nat ives f rom comp ou nders. Too-low reimbursement for generic injectables also may limit pharmaceutical industry interest in producing these therapies, leading to shortages and greater reliance on compounders. K V Pharma weighed in that the NECC case illustrates FDA’s error in permitting compounders to continue to produce hydroxyprogesterone to prevent premature births after approving KV’s Makena. State health agencies and insurers have been opting for the less costly compounded version, but now may shift to the KV product to avoid exposure to possibly unsafe compounded medicines. There will be f urther debate ove r how muc h a d d e d le g a l authority FDA needs to deal with compounders. Some agency critics complained that FDA did not make full use of its existing legal authority to regulate NECC follow i ng i n it i a l u n s at i sf ac tor y inspections. Republicans generally oppose giving the agency stronger legal powers, and compounding pharmacies claim they are sufficiently regulated by state licensing boards. Pharmacists object to the Markey bill for imposing too-broad FDA regulation of compounding that could block patient access to needed medicines and further overtax FDA. It’s a costversus-safety issue, and a challenge to find a compromise that passes muster. ◆ December 2012 www.biopharminternational.com BioPharm International 11 Employment Survey Job Security in a Changing Biopharma Environment Amy Ritter Results from the 2012 employment survey. R eader responses to BioPharm International’s annual employment survey suggest that the mergers and acquisitions that dominated the news a few years ago are slowing. However, bio/pharmaceutical companies still face pressure to run leaner businesses and to see a better return on investment from their R&D divisions. Development of large-molecule therapeutics is an area of intense interest for large pharma and small biotech companies, but biologics are expensive to develop, which affects companies’ bottom lines. In addition, bio/pharmaceutical companies face pricing pressure from cost-conscious payers and from developing countries determined to hold the line on drug prices. The industry continues to adapt to this challenging business environment, and 12 BioPharm International www.biopharminternational.com December 2012 these challenges cannot help but affect pharma employees. When readers were asked how secure they felt in their positions, fewer than last year said they felt less secure—33%, compared with 38% in 2011. While this seems encouraging, readers did not say they felt more secure. Instead, the largest group of respondents (49%) said they felt about the same as last year. It seems, then, that pharma employees are becoming accustomed to the new, more fluid business environment. Is insecurity becoming the new normal? Perhaps, but most readers felt confident they would be able to find a new job if they had to, and readers continue to derive satisfaction from the intellectual stimulation and challenging projects associated with their jobs. The following pages highlight key results from the survey. 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! Employment Survey Has your job become more global in nature (i.e., more offices/sites and/or partners in other countries) over the past two years? How secure do you feel in your job compared with last year? I feel more secure now I feel less secure now No change 2011 23.3% 2012 48.5% 38.6% 30.6% 18.6% 58.5% 32.9% 38.1% 10.9% More global interactions If it was necessary for you to change jobs this year, how would you assess the job market? 15.6% 45.1% Fewer global interactions About the same It would be straightforward to find a job Has having offices/partners in other countries made your job: comparable to the one I have now. It would take a while, but I would be able to find a job comparable 38.9% to the one I have now. 15.9% It would be straightforward to find a job, but it probably wouldn’t 23.4% I would have to search hard, and be prepared to take what I could 53.4% be as good as the one I have now. get. Within the past year, has your workload increased, decreased, or stayed the same? 7.8% 2011 2012 More difficult Less difficult No different than working with local partners 71.1% 57.1% How do you communicate with other offices or partners in other countries (check all that apply)? Increased Decreased Stayed the same 98.4% Teleconference 81.9% In-person visits 56% Email 35.3% 24.1% 7.6% 4.8% Skype or other streaming video 24.9% Employment Survey This is the main reason I come to work In the past two years, have you been through a merger, acquisition, downsizing or restructuring? 39.8% Intellectual stimulation Challenging projects 38.9% YES NO 2012 2011 $ MEDIAN SALARIE US (USD) Canada (CAD) 44.6% 55.4% 57.7% 42.3% Salary Professional advancement 110,000 72,500 (72,535 USD) What is your prediction for your company’s business prospects in the coming year? Business will improve. Business will decline. No significant change expected. 28.6% 32.1% 30.3% 50.2% 16.3% 33.6% I would change jobs for this alone. In your view, what is the general outlook for the bio/pharmaceutical industry in the short- and long-term? 51.8% Business will improve. 11.2% 14.9% *Due to rounding, some percentages may not add up to 100%. Some questions allowed multiple answers. 3.6% 2.6% 0.3% 15.5% Business will decline. Business will improve overseas, but not domestically. Business will improve domestically, but not overseas. Business will decline domestically, but not overseas. Business will decline overseas, but not domestically. No significant change expected. Job security CORPORATE CAPABILITIES www.biopharminternational.com Althea Technologies, Inc. Althea Technologies, Inc. Althea is a contract developer and manufacturer of biopharmaceutical and injectable products, with fully integrated product development and cGMP manufacturing expertise to support client projects from pre-clinical development through commercial supply. ence with complex formulations, including liposomes, nanoparticles, conjugates, and adjuvants, and are committed to the delivery of stable and high quality product. If your product requires the additional stability of lyophilization, we can lyophilize your product in our facility. Biologics Manufacturing—Recombinant Protein & Plasmid Althea can take your microbial-expressed product from cell bank to final filled product. Because of Althea’s seamless integration, your product will be smoothly transferred from Process Development, where our group will develop processes to manufacture the highest quality product, to our cGMP manufacturing facility. Clinical Packaging & Distribution—The Complete Package™ Althea can now provide The Complete Package™ through our alliance with Sherpa Clinical Packaging. Sherpa is located immediately adjacent to Althea and provides Clinical Trial Material management services including customized clinical kit design, clinical labeling, secondary packaging, storage and distribution to clinical sites worldwide. Sherpa offers the packaging and cold chain capabilities you need, while providing the flexibility and customer service you demand. Protein Analytics—Analytical Method Development & Product Characterization Offering core services of method development and validation, product characterization, comparability studies, reference standard qualification, and stability and release testing, our scientists take the approach that every molecule is unique, and requires multiple methods for characterization. ALTHEA TECHNOLOGIES, INC. 11040 Roselle St. San Diego, CA 92121 Crystalomics®—Protein Delivery Technology Althea’s Crystalomics® technology allows proteins and other large molecules to be formulated as crystals. Products formulated with Crystalomics® can be delivered at a high concentration in a small volume, and can have extended release of a week or more, ensuring increased stability and patient safety. As one of the few fully-integrated development and manufacturing service providers in the industry, Althea can consolidate many of the steps in your supply chain, from drug substance to final product. When you partner with Althea, you reduce the time, cost, and risk associated with moving product across multiple service providers. Contact us today to discuss how Althea can drive value to your drug development and manufacturing program. www.ALTHEATECH.com TELEPHONE 858.882.0123 FAX 858.882.0133 EMAIL info@altheatech.com WEBSITE www.altheatech.com 16 Aseptic Fill & Finish—Vials, Syringes, Lyophilization We can provide you with a reliable supply of final product in vials or prefilled syringes for every stage of development and commercialization. We have tremendous experi- BioPharm International December 2012 ADVERTORIAL 'SPNDPODFQU UPDPNQMFUJPO PROCESS DEVELOPMENT Cell Line Development Upstream Development Downstream Development Quality by Design DOE Based Development ANALYTICAL METHOD DEVELOPMENT Method Development & Validation Protein Characterization Comparability Studies Reference Standard Qualification cGMP PROTEIN & PLASMID PRODUCTION Cell Banking Preclinical Tox Clinical Supply Commercial Supply 1.888.4ALTHEA | altheatech.com ASEPTIC FILL & FINISH Clinical Supply Commercial Supply Vial & Syringe Filling Lyophilization ICH Stability Testing Clinical Packaging & Distribution HI-CONCENTRATION PROTEINS Up to 350 mgs/mL SC mAb Formulations Sustained Release Low Viscosity Membrane Scale-Down A New Scale-Down Membrane Adsorber Device for Process Development and Validation Nathalie Frau, Martin Leuthold, Amit Mehta, Kome (Kevin) Shomglin, and Rene Faber ABSTRACT Anion exchange membrane chromatography (AEX) is an attractive alternative to flow-through anion exchange column chromatography. Replacing AEX column chromatography with AEX membrane chromatography provides similar output but at a much higher load density, usually greater than 10 kg/L of membrane. The commercially available scale-down model, Sartobind nano, which has a 1 mL membrane volume, requires a significant amount of material for process development and validation whereas a relatively small amount of material is typically available during early clinical development. To overcome this limitation, an ultra scale-down device, Sartobind pico, was developed to reduce material consumption and validation cost. In this article, the development of the new ultra scale-down device is detailed and scalability to Sartobind nano and to a large-scale capsule are demonstrated. Studies using model proteins and industrially relevant monoclonal antibody feedstock are described. The new ultra scale-down device, Sartobind pico, enables process development, characterization, and validation with scalability to large-scale membrane chromatography devices while reducing sample consumption, time, and cost. Nathalie Frau, PhD*, is a senior scientist in R&D process technologies at Sartorius Stedim North America, Bohemia NY; Martin Leuthold, PhD, is a scientist in R&D product development at Sartorius Stedim Biotech, Goettingen, Germany; Amit Mehta, PhD, is a senior engineer in purification development and Kome (Kevin) Shomglin, PhD, is a senior research associate in purfication development at Genentech, South San Francisco, CA; and Rene Faber, PhD, is vice-president, R&D process technologies at Sartorius Stedim, North America, Bohemia NY. *To whom correspondence should be addressed, nathalie.frau@sartorius-stedim.com. PEER-REVIEWED Article submitted: Jul. 23, 2012. Article accepted: Aug. 3, 2012. 18 A nion-exchange (AEX) membrane chromatog raphy is an attractive technology for monoclonal antibody (mAb) purification because of advantages such as elimination of column packing and unpacking, higher throughput, smaller plant foot pr int, and considerably less buffer consumption. Compared with AEX resins, which are typically loaded to approximately 100 g/L, AEX membranes can provide orders of magnitude higher loading capacity in flow-through mode w it h adequate impu r it y remova l. For example, Zhou et al. reported greater than 3000 g/m 2 or 10.9 kg/L load capacity with > 5 log reduction value (LRV) for four different model viruses (1). In another study, Zhou et al. showed that a similar LRV for X-MuLV could be obtained at a load BioPharm International www.biopharminternational.com December 2012 capacity of 13 kg/L and at flow rate of 600 cm/hr (2). Glynn et al. recently described the evolution of Pfizer’s antibody purification process from three columns to two by replacing the resin-based AEX chromatography step with a membrane adsorber and increasing the load capacity of this step by a factor of 100 (3). The removal of process-related impurities with AEX membrane adsorbers at high load capacity and high flow rate has also been published by Arunakumari et al. (4). Lately, the authors demonstrated virus removal by membrane adsorbers with a LRV greater than 4.5 and 4.4 for X-MuLV and MMV, respectively, at 20 kg/L mAb load capacity (5). Mehta et al. showed that purity and product quality comparable to traditional three-column affinity processes can be achieved with a novel YOUR PROJECT IS OUR FOCUS. From consulting and development to cGMP production for clinical studies and market supply, we customize our services to bring your processes to fruition. Corporate Project Management Cell Line Development Bioprocess Development API Production Formulation, Fill & Finish Quality Control Regulatory Affairs Quality Assurance Rentschler Biotechnologie GmbH Erwin-Rentschler-Straße 21 88471 Laupheim / Germany 0HONEs%-AILINFO RENTSCHLERDE www.rentschler.de ALL FIGURES ARE COURTESY OF THE AUTHORS Membrane Scale-Down process using a nonaffinity capture step and membrane-based technologies such as AEX membrane adsorbers and high performance tangential flow filtration (6). It is thus well documented in the literature that an AEX membrane adsorber is a powerful alternative to column chromatography and can facilitate development of new purification strategies for downstream processing in the biopharmaceutical industry (7). However, the high load capacity achieved with membrane adsorbers in the flow-through mode implies the need for a significant amount of material for process development with laboratory-scale devices. For example, a load capacity of 10 kg/L means that 10 g of material is required for each experiment with a 1 mL laboratoryscale device. High material consumption can be a limiting factor, particularly during early stages of drug development where relatively small amount of material is typically available. Reducing the virus validation cost by minimizing the amount of virus spike required is also of significant interest. To overcome these limitations, a new u lt r a s c a le - dow n me mbr a ne ad s or b e r device, Sartobind pico (Sartorius Stedim Biotech GmbH, G öt t i ngen, G er ma ny), with a membrane volume of 0.08 mL has been developed. The 12.5-fold lower membrane volume than the current laboratory-scale device, 1 mL Sartobind Nano, significantly minimizes feedstock and virus spike requirements for development, characterization, and validation studies. The performance of this device was evaluated using model molecules and industrially relevant mAb feedstock and was compared with the current scale-down device, Sartobind nano. Data demonstrating the scalability of the new ultra scale-down device to a manufacturing-scale device are also presented. MATERIALS AND METHODS Devices Sartobind pico, the new scale-down device was provided by Sartorius Stedim Biotech GmbH, Göttingen, Germany. The device consists of 15 membrane layers with polypropylene sealing rings every 3 layers, and is assembled into a molded polypropylene housing with luer lock connectors to enable easy connection to a liquid chroma- 20 Table I: Key attributes of Sartobind pico and Sartobind nano. Sartobind nano Sartobind pico Bed height (mm) 4 4 Membrane volume (mL) 1 0.08 Housing materials Polypropylene Polypropylene Connectors Luer-Lock Luer-Lock Flow path Radial Axial Figure 1: A. Sartobind pico 0.08 mL. B. Sartobind nano 1 mL. Figure 2: Sartobind pico device design. Luer lock Molded polypropylene housing Stacked membrane design (5 x 3 layers) tography system (see Figures 1 and 2). The bed height of 4 mm is similar across the entire Sartobind SingleSep family and the frontal surface area of 20 mm² gives pico a membrane volume of 0.08 mL. Sartobind nano, (Sartorius Stedim Biotech GmbH, Göttingen, Germany) with 15 layers, 36.4 cm 2 total surface area, and 1 mL membrane volume was used as a reference device (see Figure 1). The Sartobind nano has a radial flow and is constructed in the same way BioPharm International www.biopharminternational.com December 2012 Setting New Standards: Platform Solutions for High Quality Analytics of Process Contaminants and Impurities 0N-DEMAND WEBCAST Register free at www.biopharminternational.com/platform EVENT OVERVIEW: Monitoring biopharmaceutical product quality requires uncompromised analytics where high quality data combined with a fast time to result enables efficient development and manufacturing processes. This webinar will describe an integrated platform of rapid, highly sensitive in-process testing methods that deliver industry-leading solutions for contaminant and impurity detection and quantitation. The methods include easy-to-use molecular technologies, including quantitative PCR, DNA sequencing, and automated sample preparation that can be applied from cell line development to formulation. Specific topics to be covered are: In-process monitoring during cell-culture manufacturing Using an integrated sample-to-results detection system for detection of Mycoplasma, MMV and Vesivirus High-throughput residual host cell DNA quantitation An immunoassay platform for analysis of residual proteins Key Learning Objectives: Understand how and where rapid molecular methods for highly sensitive contaminant and impurity testing can be applied Learn strategies for using in-process monitoring during cell-culture manufacturing as an early warning signal of a contamination event Gain insight into an integrated analytical testing platform that can help increase throughput throughout the workflow Who Should Attend: This program is intended for scientists working on or interested in learning more about analytical methods for pharmaceutical contaminant and impurity testing in such areas as: Speaker: Analytical Testing Wesley Straub Process Development Senior Technical Product Specialist, Pharmaceutical Analytics Process Engineering Life Technologies Automation Engineering Quality Control Moderator: Angie Drakulich Managing Editor BioPharm International Presented by Manufacturing Science Technical Services/Technology Development Sponsored by For questions, contact Sara Barschdorf at sbarschdorf@advanstar.com Membrane Scale-Down Figure 3: Normalized flow (MV/min) for Sartobind pico and nano devices as a function of inlet pressure. MV is membrane volume. 80 70 Flow [MV/min] 60 50 40 30 20 10 0 0 0.5 1 1.5 2 2.5 3 3.5 Pressure [bar] Pico 1 Pico 2 Nano 1 Nano 2 as process scale SingleSep capsules, which assures direct scalability to manufacturing scale capsules (7–11). The key attributes of Sartobind pico and Sartobind nano are summarized in Table I . The Sartobind SingleSep 10” capsule with a membrane volume of 180 mL was used to further confirm scalability. The devices were assembled with a salt tolerant AEX membrane, Sartobind STIC PA, consisting of a polyallylamine ligand covalently coupled to the cellulose membrane matrix (12). Equipment A l l l a b o r at o r y- s c a l e c h r o m at o g r ap hy experiments with mAb feedstock, model proteins, and model DNA were performed using an ÄKTA Explorer FPLC system (GE Healthcare Bio-Sciences Corp., Piscataway, NJ, USA). The devices were connected to the ÄKTA Explorer with standard tubing and luer-lock connectors. A flow rate of 10 membrane volume (MV)/min was used. Binding of endotoxin and bacteriophage molecules was performed using a separate experimental setup consisting of a peristaltic pump (Watson Marlow 302S), which allowed proper cleaning of the system. To determine flow rates, membrane adsorber devices were connected to a pressure vessel filled with buffer or protein solution. The filtrate volume was monitored using a balance and the flow rates for different pressures were calculated up to an inlet pressure of 3 bar. 22 Model systems Bovine serum albumin (BSA, Lot 50121326) was purchased f rom K raeber GmbH & Co. and salmon sperm DNA (DNA, Lot 8087) from Biomol. The protein throughput wa s deter m i ned usi ng γ -g lobu l i n (Sigma, γ−globulin from bovine blood, Lot STB0227K9). Endotoxin from Escherishia coli (Lonza LPS E. coli 055:B5 N185 Lot 0 0 0 010 0778) w a s u s e d a s s t a n d a r d . Bacteriophage ΦX174 (ATCC 13706-B1) was produced in a 50 L disposable bioreactor using the E. coli (ATCC 13706) expression system. Subsequently, phage was purified, concentrated, and sterile filtered by several steps including a depth filtration cascade, crossflow filtration, precipitation with polyethylene glycol, and centrifugation. MAb feedstock The mAb feedstock was obtained from pilotscale batches produced at Genentech (a member of the Roche Group). It was expressed in mammalian cells and clarified to remove insoluble impurities. The mAb was processed through a protein A chromatography step and further purified using a cation-exchange chromatography step. Protein concentration was approximately 11 g/L. METHODS Dynamic binding capacity Each device was sanitized with 1 N NaOH for 30 min at 10 M V/min followed by equilibration with 150 MV binding buffer composed of 150 mM NaCl in 20 mM Tris/HCl pH 7.3 ± 0.1, conductivity 16 mS/ cm. 150 MV of 1 g/L BSA in binding buffer or 0.1 g/L DNA in binding buffer were loaded. All solutions used were prefiltered with a 0.2 μm membrane filter. All steps were performed at flow rate of 10 MV/min. Breakthrough c ur ves were recorded by measuring the extinction at 280 nm (protein) and 260 nm (DNA) using the ÄKTA Explorer. To compare different devices the void volume of the experimental setup was determined by injection of acetone (2 %). The dynamic binding capacity at 10% breakthrough was calculated as shown in Equation 1, DBC = BioPharm International www.biopharminternational.com December 2012 (V10% – Vv)* Ci VM [Eq. 1] Membrane Scale-Down Protein throughput Each membrane adsorber device was sanitized with 1 N NaOH for 30 min at 10 MV/ min followed by equilibration with 100 MV binding buffer composed of 150 mM NaCl in 20 mM Tris/HCl pH 7.3 ± 0.1, conductivity 16 mS/cm. Protein throughput was determined using the pressure vessel filled with a solution of 20 g/L γ-globulin in binding buffer was used to determine the protein throughput with the membrane adsorber devices. The filtrate volume up to 1000 MV was monitored at a constant pressure of 3 bar using a balance. Chinese hamster ovary proteins clearance Chinese hamster ovary proteins (CHOP) clearance was determined using industrially relevant mAb feedstock. Before loading the MAb feedstock onto the membrane adsorber, the membrane was equilibrated with 10 MV of 50 mM Tris buffer at the appropriate pH. The conductivity of this buffer was adjusted by altering the concentration of sodium acetate. After equilibration, the mAb feedstock was loaded onto the devices to a targeted load density of 10 kg mAb/L of membrane at a flow rate of 10 MV/min. Pool fractions were collected during the experiment and analyzed for CHOP concentration. Determination of log reduction value of bacteriophages Equipment and membrane devices were sanitized with 1 M sodium hydroxide for 30 minutes. Membrane devices were further equilibrated with 300 MV of binding buffer. The ΦX174 phage solution with a titer of 1.5x107 PFU/mL was prepared and loaded onto the devices at a flow rate of 10 MV/ min. Flow-through fractions were collected after 100 and 150 MV of load for quantitative analysis. Endotoxin removal Pump, tubing, and devices were treated with 1 M sodium hydroxide for 30 minutes at room temperature and at a flow rate of 10 MV/min before performing the experiment. Compatible vessels and materials Figure 4: Filtrate flow rate at 3 bar constant initial pressure during loading of 20 g/L γ-globulin protein solution. MV is membrane volume. 35 Flow [MV/(min*bar)] where V10% is volume loaded at 10% breakthrough, Vv is void volume, Vm is membrane volume, and ci is initial concentration. 30 25 20 15 10 5 0 0 5 10 Load density [kg/L] Pico 1 Pico 2 Nano 1 15 20 Nano 2 were heated at 200 ∘C for 4 hours to destroy naturally occurring endotoxins. After sufficient rinsing with reverse osmosis water, the equilibration was performed with 300 MV of binding buffer. 150 MV of endotoxin in binding buffer were loaded to the membrane at a flow rate of 10 MV/min. The flowthrough was divided into fractions of 50 MV each and was analyzed to determine the endotoxin level. ASSAYS CHOP quantification An ELISA was used for CHOP quantification. Samples containing CHOP were incubated in the wells, followed by incubation w ith anti- CHOP antibodies conjugated with horseradish peroxidase (HRP). The HRP enzymatic activity was detected with o-phenylenediamine, and the CHOP was quantified by reading absorbance at 490 nm in a microtiter plate reader. Based on the principles of sandwich ELISA, the concentration of peroxidase corresponded to the CHOP concentration. The assay range for the ELISA was typically 10–320 ng/mL, with intra-assay variability of approximately 10%. CHOP values were reported in units of ng/mL. CHOP values could be divided by the mAb concentration and the results reported in units of PPM (parts per million; ng of CHOP/mg of mAb). December 2012 www.biopharminternational.com BioPharm International 23 Membrane Scale-Down Figure 5: Bovine serum albumin breakthrough curves for pico, nano, and 10” devices. MV is membrane volume. The LRV was calculated using Equation 3, LRV = log10 C0 CFT 1 [Eq. 3] where c0 was the titer of the initial solution and cFT the titer in the flow-through fraction. 0.8 C/C0 0.6 0.4 0.2 0 0 20 40 60 80 100 Load Volume [MV] Pico 1 Pico 2 Nano 1 Nano 2 10” capsule 1 10” capsule 2 Bacteriophage ΦX174 quantification Host organism E. coli was used for the detection of infectious ΦX174 phage particles. E. coli cells were incubated on agar plates (Soybean- Casein Digest Agar Medium– Trypticase Soy Broth 211043), which served as a base layer with nutrients. E. coli cells multiplied rapidly and formed a bacterial lawn. Phage particles infect the cells, causing the lysis of E. coli host cells and producing single circular, nonturbid areas called plaques in the bacterial lawn. Each plaque represents the lysis of a phage-infected bacterial culture and is designated as a plaque-forming unit (pfu), and used to quantitate the number of infective phage particles in the culture. Plaques must be clearly defined and samples were then diluted several times (1:10) depending on the phage concentration. During the study, 150 μL of the host cell solution (optical density 2–6) was mixed with 150 μL of sample and top agar (1.3% Tryptikase Soy Agar BD 211043) and the mixture was then distributed to agar plates (4% Tryptikase Soy Agar BD 211043 in 90 mm petri dishes) and incubated for 18 to 24 hours at 37 ∘C. Plaque forming units were counted and the titer of the sample in PFU/mL (plaque forming units per mL) was calculated using Equation 2, Titer = P E D VSample [Eq. 2] where P is the number of plaques of all countable dilutions, E is the sum of emphasis, D is the lowest evaluated dilution, and VSample is the sample volume. 24 Endotoxin quantification The endotoxin level was measured by the kinetic chromogenic method test according to t he manufac t urer’s inst r uc t ions (Limulus Amebocyte Lysate Chromogen, Cha rles R iver endosa fe E ndoch rome -K R1710K, Lot A4992L 10/2012). The quantification principle is based on coloration caused by the contact of a sample containing endotoxin with a mixture of lysate and chromogenic substrate. A β -glucan blo c ke r w a s a d d e d ( L o n z a N19 0 L o t 0 0 0 0132199 01/11). D ur ing t he 1-hour incubation the extinction coefficient was measured continuously at 405 nm using a temperature controlled (37 ∘C) plate reader ( Tecan Saf ire). T he react ion rate var ies with endotoxin level and the samples were quantified for endotoxin by comparing t he results w it h t he calibrat ion ser ies. The detection limit of the assay was 0.012 EU/mL. LRV was calculated similarly to phage quantification by measuring the endotoxin level of the initial solution El0 and t he level of endotox in in t he collected flowthrough fractions (ElFT) using Equation 4 . LRV = log10 EI0 EIFT [Eq. 4] RESULTS Flow rate and protein throughput Device geometry must allow for linear scalability through the entire device size range. Pressure flow curves were generated with the axial flow Sartobind pico and radial f low Sartobind nano devices with data shown in Figure 3. The normalized flow rate (membrane volume (MV)/minute) increased linearly with the increasing inlet pressure and the flow rates were comparable, suggesting effective flow distribution and efficient utilization of membrane area with both pico and nano devices. For a typical polishing application with an AEX membrane adsorber, the load capac- BioPharm International www.biopharminternational.com December 2012 Membrane Scale-Down Table II: Dynamic binding capacity (DBC) at 10% breakthrough using bovine serum albumin (BSA) and DNA model molecules. BSA is bovine serum albumin. Membrane volume (mL) 10% DBC BSA (g/L) 10 % DBC DNA (g/L) Pico 1 0.08 55.83 9.06 Pico 2 0.08 50.78 9.43 Pico 3 0.08 50.78 8.94 Pico 4 0.08 48.25 9.06 51.41 9.12 Device Average (Pico) Nano 1 1 53.54 8.94 Nano 2 1 49.11 9.78 51.32 8.52 Average (Nano) 10" 180 51.84 8.02 10" 180 52.89 7.51 52.42 7.70 Average (10”) Table III: Log reduction value of bacteriophage φX174 with Sartobind pico and nano devices. MV is membrane volume. Load volume (MV) Pico 1 Pico 2 Pico 3 Pico 4 Nano 1 Nano 2 100 5.4 5.1 5.1 5.3 5.2 5.5 150 5.5 4.9 4.8 5.1 5.3 5.3 Average 5.4 5.0 5.0 5.2 5.3 5.4 ity is very high, exceeding 10 kg of protein feedstock per liter of membrane volume and can thus present the risk of membrane fouling. To assess fouling as a function of load capacity, the Sartobind pico and Sartobind nano devices were loaded with a 20 g/L γ-globulin solution to a load capacity of 20 kg/L at a constant inlet pressure of 3 bar. As seen in Figure 4, while slightly higher flow decay was observed with the pico device, the overall flow decay was minimal with the two devices thus demonstrating the absence of significant membrane fouling at high load density. Characterization of membrane adsorber devices using model systems Chromatography media are usually characterized using model molec ules, w ith dynamic binding capacity and impurity clearance reported at specific process conditions. The dynamic binding capacity for Sartobind STIC-PA was determined using bovine serum albumin (BSA) and DNA, and impurity clearance was evaluated using DNA, endotoxin, and bacteriophage. Dynamic binding capacity: The dynamic binding capacit y at 10% breakthrough was measured for the Sartobind pico, the Sartobind nano, and the Sartobind SingleSep 10” capsule using BSA and DNA model systems. All devices were assembled with STIC-PA membranes. The breakthrough curves for the three devices are shown in Figures 5 and 6 for BSA and DNA, respectively. The breakthrough curves are similar for all devices suggesting consistent flow distribution and efficient utilization of the membrane binding sites at the three scales. Table II shows the average BSA and DNA dynamic binding capacity values for several Sartobind pico, nano and 10” devices. At 10% breakthrough, the difference in dynamic binding capacity for all three devices was insignificant. The consistent dynamic binding capacity with BSA and DNA supports a linear scalability from 0.08 mL axial flow pico device to 180 mL radial flow SingleSep 10” capsule. December 2012 www.biopharminternational.com BioPharm International 25 Membrane Scale-Down Table IV: Endotoxin removal (log reduction value) at pH 7.3 in buffer containing 150 mM NaCl with Sartobind pico and nano devices. MV is membrane volume. Load volume (MV) Pico 1 Pico 2 Pico 3 Pico 4 Nano 1 Nano 2 50 > 3.96 > 3.96 > 2.92 > 3.96 > 3.96 > 3.96 150 > 3.96 > 3.96 > 3.96 > 3.96 > 3.96 > 3.96 150 > 3.96 > 3.96 > 3.96 > 3.96 > 3.96 > 3.96 Figure 6: DNA breakthrough curves for pico, nano, and 10” devices. MV is membrane volume. 1 C/C0 0.8 0.6 0.4 0.2 0 0 20 40 60 80 100 Load Volume [MV] Pico 1 Pico 2 Nano 1 Nano 2 10” capsule 1 10” capsule 2 Removal of bacteriophage: Pathogen clearance was evaluated using the bacteriophage ΦX174, serving as a surrogate for mouse minute virus (MMV), which is typically used as a model virus for virus validation studies. Both ΦX174 (26-33 nm diameter) and MMV (20 nm diameter) are small nonenveloped DNA viruses with an isoelectric point of around 6.7–7.0 and 6.2 respectively (13). At pH > 7, both ΦX174 and MMV are mainly negatively charged and expected to bind to positively charged AEX chromatography membranes, resulting in their clearance from protein feedstock through electrostatic interactions. To compare clearance between Sartobind pico and Sartobind nano, the same ratio of ΦX174 to membrane volume was loaded. Processscale capsules were not tested because of the large amount of phage material required. Two flow-through fractions were collected with each pico and nano device, and the LRV was evaluated by comparing the phage titers of the fractions with the load solution. As shown in Table III, similar LRVs were obtained at a load of 100 and 150 MV of phage-spiked buffer, demonstrating linear scalability between the devices. 26 Removal of endotoxin: Endotoxins are lipopolysaccharides found in the outer membrane of various gram negative bacteria, can be present as different forms of micelles and vesicles, and are generally strongly negatively charged. Because of their ability to elicit immunogenic responses in humans, endotoxins must be removed to typically < 0.25 Endotoxin Units per milliliter (EU/mL) where EU is the unit of measurement for endotoxin activity (USP <29>). Table IV shows the results for endotoxin removal with Sartobind pico and nano devices at pH 7.3 in a buffer containing 150 mM sodium chloride. The concentration of endotoxin in the load was 108 EU/ mL, which is significantly higher than the concentration of endotoxin typically found in any in-process pools. Three fractions were collected from the flow-through at loading volumes of 50, 100, and 150 MV. All flowthrough fractions had an endotoxin concentration below the detection limit of 0.012 EU/ mL resulting in a LRV > 3.96 except one fraction at 50 MV with the pico device. However, subsequent fractions at higher load volumes with the same pico device provided LRV > 3.96 which suggests that the anomalous reading at 50 MV was likely due to an assay error or sample contamination. Based on the load volumes tested, the total amount of endotoxin removal was > 1296 EU with the pico and > 16200 EU with the nano device. Significantly larger amount of endotoxin would be required in the load to saturate the membrane with the endotoxin molecules to determine and compare the breakthrough curves for both pico and nano devices. Performance of Sartobind pico with an industrially relevant mAb feedstream In a mAb purification process, AEX chromatog raphy is t y pically operated in a flow-through mode to bind trace levels of impurities such as DNA, putative viruses, BioPharm International www.biopharminternational.com December 2012 Membrane Scale-Down Figure 7: Chinese hamster ovary proteins (CHOP) breakthrough curves for Sartobind pico and nano with a mAb feedstream. MAb feedstock contained 100 ppm CHOP. Experiments were performed at pH 8.0 and 7.0 at 11 mS/cm and at a flow rate of 10 MV/min. 80 CHOP (ppm) 60 Together, we can produce mycoplasmafree cell culture media...............faster 40 20 0 0 2 4 6 Load density [kg/L] 8 pH 7, 11 mS/cm Pico pH 7, 11 mS/cm Nano pH 8, 11 mS/cm Pico pH 8, 11 mS/cm Nano endotoxins, and host cell pro teins, wh ile t he m Ab produc t flows through. The load capacity is indicated as the mass of product loaded per unit volume of chromatography membrane (kg mAb/L membrane) such that the purity level in the product pool is acceptable. To assess the performance with an industrially relevant feedstream, both pico and nano devices were loaded with an in-process mAb pool post Protein A and cation exchange chromato g r aphy s te p. S u b s e qu e nt l y, CHOP levels were monitored in the f low-through as a function of load density. The devices were loaded to 10 kg/L load densit y at two different solution conditions (pH 7.0 and 8.0 at 11 mS/ cm). CHOP clearance as a function of load density is shown in Figure 7. Comparable CHOP clearance was obtained with the pico and the nano device at both solution conditions using an industrially relevant mAb feedstock, suggesting that the Sartobind pico is scalable to the Sartobind nano device. Additionally, at pH 7.0 and 11 10 mS/cm, a load capacity \ 10 kg/L could be achieved with pool CHOP levels < 10 ppm. The CHOP clearance results are consistent with the earlier data where comparable BSA and DNA dynamic binding capacity was observed between the pico, nano, and process scale 10“ devices. Comparable clearance of endotoxin and the bacteriophage further demonstrated the scalability of Sartobind pico to the Sartobind nano. CONCLUSION It is well documented in the literature that AEX membrane adsorbers are an attractive alternative to columns for polishing applications in a flow-through mode. Because of its hydrodynamic benefits, load capacity greater than 10 k g / L of me mbra ne c a n b e achieved with membrane chromatography. Such high load densit y necessitates a significantly la rge a mou nt of protei n feedstock for process development and validation, which could be cost proh ibit ive. To overcome this limitation and also to reduce 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 Faster mycoplasma batch removal processing Typical LRV >10 for Acholeplasma laidlawii Industry leading flow rates Reduced filtration costs Integral prefilter layer for increased capacity Incorporate PROPOR MR into single-use automated systems at: www.scilog.com www.parker.com/processfiltration Europe: phone +44 (0)191 4105121 email: dhprocess@parker.com North America: toll free: +1 877 784 2234 email: dhpsales.na@parker.com December 2012 www.biopharminternational.com BioPharm International 27 Membrane Scale-Down validation cost particularly for virus spiking studies, an ultra scale-down device, Sartobind pico, having a membrane volume of 0.0 8 mL was developed. Using mo de l mole c u le s a nd a n i ndu st r ia l ly relevant mAb feedstock, Sartobind pico was compared to the existing commercial scale- down dev ice Sartobind nano. BSA and DNA breakthrough curves, CHOP, b a c te r io p h a ge, a nd e nd o t ox i n c le a rance data demonstrate the scalability of Sartobind pico to the Sartobind nano. The new scale-down pico device will facilitate the development of flow-through polishing applications for recombinant proteins and monoclonal antibodies by reducing the sample consumption by 10 -fold and providing substantial cost savings for process characterization and virus validation studies. REFERENCES 1. J.X. Zhou and T. Tressel, Biotechnol. Prog. 22, 341–349 (2006). 2. J.X. Zhou et al., J. Chromatogr. A 2006,1134, 66–73. 3. J. Glynn et al., “Downstream Procecessing 2010” supplement to Biopharm Int. 22, s16– s20 (2009). 4. A. Arunakumari, J. Wang, and G. Ferreira, “Advances in Process Chromatography” supplement to Biopharm Int. 22 s36–s40 (2007). 5. A. Arunakumari, J. Wang, and G. Ferreira, “Downstream Procecessing 2010” supplement to Biopharm Int. 22 s22–s26 (2009). 6. A. Mehta et al., “SBE Supplement– Bioprocessing” supplement to Chemical Engineering Progress 104, 14–20 (2008). 7. N. Fraud, Bioprocessing J. 7, 34–37 (2008). 8. A. Pastor, M. Hirai, and S. FischerFruehholz, presentation at GVC/Dechema, (Osnabrück, Germany, May, 2007). 9. U. Gottschalk, Pharma Focus Asia 7, 60–65 (2008). 10. J. Zhou et al., Biotechnol. Bioeng, 100, 488– 496 (2008). 11. Sartorius Stedim Biotech GMbH, “Scale up with Sartobind SingleSep,” Application Note, SL-4042-e07081. 12. R. Faber, Y. Yang, and U. Gottschalk, Biopharm Int. 22 (10), 11–14 (2009). 13. D. M. Strauss et al., Biotechnol. Bioeng. 104, 371–380 (2009). ◆ Call for Papers * Call for Papers * Call for Papers BioPharm International integrates the science and business of biopharmaceutical development, and manufacturing. We provide practical, peer-reviewed technical solutions to enable biopharmaceutical professionals to perform their jobs more effectively. We are currently seeking novel research articles for our peer-reviewed journal as well as manuscripts for our special issues. Submitted manuscripts should be sufficiently novel to be of interest to an experienced audience. Articles should be data driven and provide sufficient technical detail to support the main thesis or should offer a novel synthesis of existing data. Topics should be timely and useful and should focus on the development of peptides, monoclonal antibodies, fusion proteins, other therapeutic proteins, nucleic acids, vaccines, cells for cell therapy, and any other class of biotechnologically generated molecular class. For peer-reviewed papers, members of BioPharm International’s Editorial Advisory Board and other industry experts review manuscripts on technical and regulatory topics. The review process is double-blind. Manuscripts are reviewed on a rolling basis. Our single-themed issues, which include literature reviews, and tutorials, cover protein characterization, biopharmaceutical manufacturing trends, vaccines, outsourcing, expression systems, and single-use technologies and facilities. BioPharm International readers are involved in product and process development, manufacturing, quality control/quality assurance, analytical technologies, regulatory affairs, plant and project engineering and design, and corporate management for the entire scope of biopharmaceutical products, including therapeutic peptides, proteins, nucleic acids, and cells for cell therapies and regenerative medicine, as well as both therapeutic and prophylactic vaccines. Please visit our website, www.BiopharmInternational.com, to view our full Author Guidelines. Manuscripts may be sent to Scientific Editor Amy Ritter at aritter@advanstar.com. WWW.BIOPHARMINTERNATIONAL.COM 28 BioPharm International www.biopharminternational.com December 2012 Set Yourself Up for Success in 2013 Stay the Week! Invest in a BDP Week Experience 5- Day Pass to Maximize Learning and Networking at the Best Rate. February 25 - March 1, 2013 Hyatt Regency Huntington Beach Resort & Spa Huntington Beach, CA The Only Conference Addressing New Methods to Help You Navigate the Development, Production and Regulatory Challenges of an Emerging Wave of mAbs and Novel Molecules Methods ¯ Implement Process Validation and Continuous Verification in the New Paradigm ¯ Leverage Contract Manufacturing and Technology Transfer for Improved Bioproduction ¯ Prepare to Meet Future Demands for Analytical Technologies ¯ Control Raw Material Variability and Improve Supply Chain Transparency ¯ Mitigate Contamination Risk with Viral Safety Strategies to Meet Regulatory Standards & View the Agenda Molecules ¯ Improve Existing and Design New Upstream Processes for Today and Tomorrow ¯ Enter the New Age of Biopharma with Next Generation Downstream Processes ¯ Develop Biosimilars that Meet Speed, Cost-Efficiency and Similarity Challenges ¯ Implement Facility of the Future with Manufacturing Strategies and Lifecycle Management ¯ Streamline the Path from Development to Commercial Success for Antibody Drug Conjugates ¯ Transform Bispecific Antibody Design, Development and Production Exclusive BioPharm International Subscriber Savings! Mention This Ad (Priority Code: BDP13BI) and Save 20% off the Standard Rates www.IBCLifeSciences.com/BDPWeek Organized by: CORPORATE CAPABILITIES www.biopharminternational.com Eurofins Lancaster Laboratories, Inc. cGMP Cell Bank Manufacturing and Cell Line Characterization t cGMP manufacturing of Master and Working Cell Banks t Full cell line characterization services for testing Master, Working, and End-of-Production Cell Banks Company Description Eurofins Lancaster Laboratories is a global leader in bio/pharmaceutical laboratory services providing innovative and timely scientific solutions to streamline the drug development process. With five service models to offer, our clients can choose the most efficient and cost-effective service solution for their project. We also provide 24-hour data access via our innovative and secure online tool at LabAccess.comSM. Markets Served We provide full CMC testing services to support more than 800 virtual and large bio/pharmaceutical companies and CMOs. Biopharmaceutical Techniques Supported Biochemistry t Spectrophotometry (CD, Bradford, A280, BCA) t Electrophoresis (CE, SDS-PAGE, 2D gels, western blots, IEF) t Chromatography (AAA, peptide mapping, glycan profiling, SEC, CEX, RP-HPLC and UPLC) t Mass spectrometry (Ion trap, LC/MS/MS, ESI-TOF, MALDI-TOF, Q-TOF, TOFTOF, Orbitrap) EUROFINS LANCASTER LABORATORIES, INC. 2425 New Holland Pike PO Box 12425 Lancaster, PA 17605-2425 TELEPHONE 717.656.2300 WEBSITE www.LancasterLabsPharm. com NUMBER OF EMPLOYEES > 1500 YEAR FOUNDED 1961 30 Molecular and Cell Biology t Cell-based potency assays (Cell proliferation, cAMP, reporter gene, cell surface receptor binding, apoptosis, alkaline phosphotase induction, lentiviral transduction) t ELISA (HCP, Protein A, sandwich ELISA, competitive ELISA) t qPCR (rDNA, Viruses) t Genetic stability Major Services t Method development, optimization, cGMP qualification and validation t Drug substance and product characterization t Full stability and release programs for clinical and marketed products t Raw material and excipient testing (USP/NF, EP, JP) t Microbiology including mycoplasma, sterility, endotoxin, and particulate matter t EU release testing t R&D and cGMP cell bank manufacturing, testing and storage t Viral and molecular biology testing t Process/facilities validation—viral clearance, residual impurity testing, extractables & leachables, water testing, environmental monitoring, disinfectant efficacy, on-site sample collection t Consulting/protocol writing. Facilities A part of Eurofins’ international network of GMP laboratories, Eurofins Lancaster Laboratories has a global capacity of over 300,000 square feet within our locations in the US and Europe. All of our facilities offer cGMP-compliant laboratory services and operate under the same strict quality control program. Utilizing the same LIMS system and centralized billing, working with any of our global facilities is a seamless operation. Virology t Viral clearance studies (Infectivity assays, qPCR assays) t Viral Safety Testing (in-vitro, retrovirus, reverse transcriptase assays) BioPharm International December 2012 ADVERTORIAL The Most Flexible Contract Lab Service Provider For 50 years 5 Service Models, including Fee For Service, FTE and our award-winning Professional Scientific StaffingSM give you endless flexibility & cost-saving laboratory service options. Participating in an average of 3-4 GMP audits a week, continually earning the trust of our 800+ leading Bio/Pharmaceutical customers. And we spring for free 24/7 online raw data & reports. Visit LancasterLabsPharm.com to learn why virtually every major Pharma & Biotech company turns to us. Global Bio/Pharmaceutical Services Method Development & Validation Stability Testing and Storage Final Product Release Testing In-vitro Bioassays Raw Materials Testing Residual & Impurities Testing Facility & Process Validation Biochemistry & Chemistry Microbiology Molecular and Cell Biology Viral Clearance & Viral Safety Cell Banking Services Mycoplasma Testing Container Closures Closures for Pharmaceutical Preparations: A Review of Design and Test Considerations Design Pics/Kelly Redinger/Getty ABSTRACT Closures that form part of the container-closure system are an important component in the packaging of sterile products. Container-closures maintain the sterility of parenteral pharmaceuticals and prevent ingress of contamination when a needle is inserted into a vial. This article describes important aspects to consider in the manufacture of closures for pharmaceutical preparations, as well as the various physical, chemical, and biological assessments required to ensure that these closures are fit for purpose. P Tim Sandle, PhD, Bio Products Laboratory, Dagger Lane, Elstree, WD6 3BX, United Kingdom, tim.sandle@bpl.co.uk PEER-REVIEWED Article submitted: June 29, 2012. Article accepted: July 10, 2012. 32 a rentera l produc ts a re desig ned, for mu lated, a nd packaged to be sterile and to maintain sterility. One of the most important parts of the packaging of sterile drug products is the container-closure mechanism. This article examines the use of closures, for products intended for injection, in the pharmaceutical industry. The article considers the most important aspects relating to the manufacture of closures and the different physical and biological assessments required to ensure that the closures are “fit for purpose.” The article does not address caps or other types of seals. Closures form part of the “containerclosure system.” Container-closures function to keep the contents of pharmaceutical preparations sterile (e.g., by providing a barrier between the neck of a vial and the vial contents) and to prevent ingress of contamination into a vial once a needle is inserted (e.g., by enabling resealing of the vial after the needle is withdrawn). The closure, together with a crimp that creates the container-closure, and the vial itself form the primary packaging or packag- BioPharm International www.biopharminternational.com December 2012 ing component (i.e., the material that first envelops the product and holds it) (1). The ideal container-closure will have low permeability to air and moisture and a high resistance to aging (2). Therefore, the manufacturers and users must have confidence in the quality control and validation of closures. It is an important part of pharmaceutical manufacturing that all information on the composition and manufacturing processes for each component type must be understood. CLOSURES Pharmaceutical closures, also known as stoppers or bungs, are an important part of the final packaging of pharmaceutical preparations, particularly those that are intended to be sterile. The most commonly used type of stopper is the elastomeric closure. An elastomer is any material that is able to resume its original shape when a deforming force is removed, which is known as viscoelasticity (3). For the manufacturing of closures, the elastomer is either natural or, as is more common, a synthetic rubber, such as butyl rubber or chlorobutyl ALL FIGURES ARE COURTESY OF THE AUTHORS Container Closures rubber. The advantage of synthetic rubbers is that the materials are strongly resistant to permeation by oxygen or to water vapor (4). In terms of the specification for closures and the testing and sterilization requirements, the following documents are useful as starting points: t '%" Guidance for Industr y: Container Closure Systems for Packaging Drugs and Biologics3PDLWJMMF.%.BZ t &VSPQFBO$PNNJTTJPOGuideline on Plastic Immediate Packaging Materials (Brussels, May 2005). t USP, General Chapter <381> Elastomeric Closure for Injections. t E u ro p e a n Ph a r m a co p e i a . C h ap te r 3, Materials for Containers and Containers. 5IF '%" Code of Federal Regulations (CFR) QBSU TUJQVMBUFT UIBU DPOUBJOFSDMPsures must provide adequate protection to the product over the product shelf-life. Before using a closure in a vial or bottle with a drug product, the closure must be assessed to determine if it is suitable for use with the product that will be filled into the glass container. The pharmaceutical manufacturer should consider the following questions relating to product compatibility, in conjunction with the manufacturer of the closure: t *TUIFQSPEVDUBCTPSCFECZUIFSVCCFS t %PFT UIF SVCCFS SFBDU XJUI UIF QSPEVDU BOEMFBDIPVUJNQVSJUJFT t "U XIJDI UFNQFSBUVSF SBOHF JT CPUI DMPTVSFBOEQSPEVDUTUBCMF t )PXFGGFDUJWFJTUIFTFBMJOUFHSJUZ t 8IBUIBQQFOTXIFOUIFQSPEVDUBOETUPQQFSBSFTUPSFEUPHFUIFSPWFSUJNF Once these questions have been satisfactorily answered, the pharmaceutical manufacturer can work with the manufacturer of the closure to design the optimal closure for the vial type and product. MANUFACTURING PROCESS The manufacturing process for closures involves processing raw materials and auxiliary substances; weighing and mixing; followed by vulcanization. Vulcanization is a chemical process for converting rubber or related polymers into more durable materials via the addition of sulfur (or another equivalent curative) together with an accelerating agent such as 2-mercaptobenzothiazole; an activator, usually zinc oxide; fillers such as carbon black or limestone; antioxidants; and lubricants. Following vulcanization, molding and compressing occur. There are two types of molding: compression and injection, of which the former is the most common. Compression molding is a method of molding in which the molding material, generally preheated, is first placed in an open, heated mold cavity. The mold is closed with a top force or plug member, and pressure is applied to force the material into contact with all mold areas, while heat and pressure are maintained until the molding material has cured. Injection molding is a manufacturing process for producing parts from both thermoplastic and thermosetting plastic materials. Material is fed into a heated barrel, mixed, and forced into a mold cavity where it cools and hardens to the configuration of the mold cavity. After molding, the stages are: coating, washing, siliconization (if required, using specific, high-viscosity silicon oil), and packaging. Siliconization has several advantages in that it prevents stoppers from sticking together or onto other surfaces and can assist with the insertion of a needle through the stopper. The siliconization step is, however, a potential source of contamination. Silicone used in the preparation of rubber stoppers should meet appropriate quality control criteria and not have an adverse effect on the safety, quality, or purity of the drug product. The mixing of raw materials and auxiliary substances involves the formulation of the stopper. A stopper is typically made up of 60% rubber, 30% fillers (which protect the physical properties of the rubber) and pigments, 5% plasticizers (which provide flexibility), 5% additional chemicals including accelerators (which help to create the crosslinkages which give the stopper its strength and hardness), activators (which are a function of the efficiency of the cross-linkages), and antioxidants (which help to avoid the degradation of the rubber). There are different types of rubber, such as natural rubber (latex), isoprene rubber (a chemical copy of natural rubber), styrolbutadine rubber, ethylene propylene dyes monomers, silicone (polysiloxane) rubber, and halogenized butyl rubber. December 2012 www.biopharminternational.com BioPharm International 33 Container Closures QUALITY CONTROL OF CLOSURES A number of quality control checks are required for the manufacture and release of closures. These checks include: After the material has been mixed t 4QFDJGJD HSBWJUZ XIJDI JT UIF SBUJP PG UIF weight of the molded piece to the weight of an equal volume of water t $PMPS XIJDI DBO CF DIFDLFE BHBJOTU B color chart t %JTQFSTJPO PG B WVMDBOJ[FE TBNQMF JO SFMBtion to particle size t &YBNJOBUJPO PG UIF BTI BGUFS CVSOJOH JO comparison to a reference sample t )BSEOFTTXIJDIDBOCFNFBTVSFEJO4IPSF Units or an equivalent standard t 3IFPMPHZ PG UIF DPNQPVOE CZ DPOEVDUJOH an examination of solids under conditions in which they respond with plastic flow rather than deforming elastically in response to an applied force. This is an assessment of the force necessary to rotate the material by 1°. An important distinction is that different materials—types of rubber and formulations—have different profile and respond in different ways. Post-compression and molding The material is checked for rubber thickness and evenness. Washing process t %VSJOH UIF MPBEJOH PG TUPQQFST JOUP B washer, the quality of the water should be checked for bioburden and endotoxin using compendial methods. t 5IF MPBEJOH PG UIF XBTIFS TIPVME UBLF place in a controlled environment, usually a cleanroom, with staff appropriately gowned. Unusually for a pharmaceutical process and in keeping with cleanrooms used in the electronics industry, the cleanroom may be fitted with deionization equipment in order to avoid fibers being attracted to the rubber. Post-washing and post-siliconization After the stoppers have been washed, a number of quality control checks should be performed. Mechanical and material tests t $PNQSFTTJPO UFTU " DPNQSFTTJPO UFTU JT performed to determine the behavior of 34 materials subjected to compressive loads. Loading is usually done at a uniform rate (in/min). t )BSEOFTT t 'SBHNFOUBUJPO t 1FOFUSBCJMJUZ JF XIBU IBQQFOT XIFO a needle passes through the stopper): Assessments can be made of the insertion force, break loose force, and extrusion force. One common issue that can arise is the generation of rubber particles cut from the closures when needles are inserted, a phenomenon sometimes referred to as coring (5). t "TTFTTNFOUPGEJNFOTJPOTBOEGMFYJCJMJUZ Many material tests are conducted by testing a selection of closures using a high-speed color sensor that examines the top, bottomside surface, and inside of the closure. Physical tests t 3FTJTUBODF UP TUFSJMJ[BUJPO 5IJT SFRVJSFT consideration of two questions: how does the rubber of the stopper react to different types of sterilization, such as gamma irradiation, ethylene oxide, and steam sterilization, and does the stopper become more CSJUUMFPWFSUJNF t 1BSUJDMFUFTUJOH Chemical tests t 5FTUT GPS FYUSBDUBCMFT BOE MFBDIBCMFT & YUSBDUBCMFT BSF DIFNJDBM TVCTUBODFT that are obtained by exposing the packag ing to a var iet y of solvents under exaggerated incubation conditions of time and temperature (6). Leachables differ from extractables in that they are chemical substances that migrate under normal conditions of use from the stopper into a drug product. Leachables are, therefore, a subset of extractables; all extractables are potential leachables of toxicological concern (7). t 4JMJDPOF PJM EFUFSNJOBUJPO 5IF FGGFDU PG subvisible silicone particles should be assessed, because these can cause aggregation with proteins, and the new complex can potentially trigger an immunochemical reaction within the body of the patient receiving the drug. Biological tests t $ZUPMPHJDBMUFTUJOH BioPharm International www.biopharminternational.com December 2012 Container Closures t #JPCVSEFO BTTFTTNFOU TVDI BT <5 CFU/ stopper). Some manufacturers undertake an examination for mesophilic counts while others focus on examining for thermophilic bacteria, because such microorganisms will be the most resistant to the sterilization step. t #BDUFSJBM FOEPUPYJO UFTUJOH TVDI BT <1 &6TUPQQFS 5IF UFTUJOH PG DMPTVSFT GPS endotoxin, using the Limulus amebocyte lysate (LAL) method, is quite difficult in terms of method validation because the endotoxin challenge to the rubber surface can prove to be tricky to recover. Container-seal tests Of the different test methods described, the assessment of the container-closure is arguably the most important because it indicates whether the device is at risk from extraneous microbial contamination. Pharmaceutical containers constructed of materials such as plastic and glass must be qualified and meet USP <661> Containers and <671> ContainersPermeation standards. The user will therefore need to undertake additional tests that examine the physical seal of the closure in the vial, i.e., when the stopper is fully inserted and crimped, usually by of an aluminium band. The choice to conduct a physical test or a microbial ingress test for this purpose is a matter of debate. Some practitioners argue that the physical methods of measuring the system’s integrity are preferred because they are more reproducible, faster, less expensive, more reliable, and quantitative. Others argue that, as the objective is to ensure that the product is safe from microbial contamination, a microbial test is the only true test. Some opt to undertake both physical and microbial tests. A review of industry practices suggests that failures occur with container-closure seals for a variety of reasons (8). These failures include poor quality starting materials, an improper fit of the container-closure combination, the lack of sufficient inspection as part of batch release, insufficient process monitoring or process control, the use of unreliable manual or visual inspection techniques, the use of methods that produce subjective results, and the lack of proper process validation. The latter point is addressed through the tests described below. Physical tests include the dye test, vacuum testing, gas leakage determined using a bubble test, liquid leakage detected by atomic absorption of a copper ion tracer solution, PS B IFMJVN MFBL SBUF UFTU 0G UIFTF UIF helium leak test is one of the most widely conducted; the objective is to detect leaks by monitoring changes in headspace gas composition or changes in total headspace pressure. This test measures the rate of helium leak from the vial as well as the actual percentage of helium that is filled within the vial. Mass spectrometry can be used to measure the rate of leakage. Mass spectrometry-based leak detection is accomplished by measuring the amount of a tracer gas that escapes from the container-closure system. Tracer egress is facilitated by a pressure difference across the container-closure barrier. Alternative and novel test methods to assess container-closure integrity include the use of hygroscopic powder and nearinfrared (NIR) spectroscopy as a means of visualization. A second example is with airborne ultrasonic technology where a sound wave is directed towards the container-closure and visualized through the creation of a high-resolution image. An alternative to ultrasound is the use of a laser diode or the utilization of high- voltage technology. These new techniques have the advantage of being non-destructive and they allow for a larger proportion of the batch to be tested, which increases the level of confidence in the integrity of the seal. These techniques are also more accurate in allowing identification of small pinholes, micro cracks and seal imperfections that cannot visually be seen. 8JUI NJDSPCJPMPHJDBM UFTUJOH B TUFSJMity test of the end product or a microbial ingress test can be considered. The sterility test is unsuitable because the test will only detect viable microorganisms present at the time of the test and those that are capable of growth within the culture media used. The microbial ingress test involves direct microbial challenge and is, therefore, a more robust test. The objective is to detect microbial ingress based on 1) the probability that the challenge microorganisms can find a container-closure leak, 2) the ability of the microorganisms to traverse the leak, and 3) the capability of the microorganisms to grow in the internal container environment. December 2012 www.biopharminternational.com BioPharm International 35 Container Closures The microbial ingress test can be performed in different ways. One of the key criteria is the selection of the microorganisms. It is more common to use two different microorganisms of different sizes and with different methods of motility. For example, Brevundimonas diminuta, a very small bacterium, and Escherichia coli, a bacterium with a relatively powerful motility, are often used in combination (10). The complexity with the test relates to achieving a sufficiently high microbial population. To conduct a microbial challenge test, vials are filled with a microbiological growth medium before stoppering and crimping, and are immersed in a 35 °C bath containing magnesium ion as well as 8 to 10 logs of viable bacterial cells for 24 hours. The test units are then incubated at 35 °C for 7 or 14 days. Microbial ingress is detected by turbidity and plating on blood agar. The described tests, or a selection thereof, should ensure that the integrity is verified over the product’s shelf-life, simulating the stresses the product will be subjected to, including sterilization, handling, and storage conditions. The tests, therefore, need to be made more rigorous in order to simulate “real life” events, for example by exposing test vials to stresses of temperature and pressure conditions, which the vials are subjected to when being transported for distribution and sales. The level of confidence is increased if three different batches are assessed. Another option is to assess vials as part of a stability trial program, which includes a time point at the end of the shelf-life. Packaging After packaging, a selection of bags should be examined for tears as a part of the quality control assessment. The placement of the stoppers into the packaging should be underUBLFO XJUIJO BO *40 $MBTT &6 (.1 (SBEF C cleanroom for standard stoppers and in an *40 $MBTT &6 (.1 (SBEF " FOWJSPONFOU for ready-to-sterilize or ready-to-use stoppers. Sterilization Closures are typically sterilized by one of two methods: steam sterilization using autoclaves and gamma irradiation. It should be noted that not all types of stoppers can be sterilized by gamma irradiation because the 36 rubber of the stopper will become brittle from the generation of free radicals in the polymeric materials (11). The sterilization of stoppers also requires the sterilization device to be subject to the standard tests including thermometric studies and biological indicators for steam sterilization devices and dosimeters for gamma irradiation. CONCLUSION The container-closure system is an essential par t of the f inal presentation of a pharmaceutical product. It defines the closure, protection, and functionality of a container while ensuring the safety and quality of the drug product over the product shelf life. This article has addressed the important considerations for closures: the “rubber” stoppers inserted into vials of products and sealed in place. The article has focused upon the important tests, control measures, and essential aspects for ensuring that the product, in its final packaging, is fit-for-purpose prior to the administration of the drug. REFERENCES 1. L. Solomun et al., J. Pharm. Biomed. Anal. 48 (3), 744–8 (2008). 2. W. Curry et al., AAPS PharmSciTech. 11 (4), 1572–9 (2010). 3. USP 32–NF 27 (Rockville, MD, 2009), pp. 4133– 4140 . 4. E.J. Smith and R.J. Nash, “Elastomeric Closures for Parenterals,” in Pharmaceutical Dosage Forms: Parenteral Medications, K.E. Avis, H.A. Lieberman, L. Lachman Eds. (Marcel Dekker, New York, 1st ed., 1992), pp. 445–507. 5. G.D. Chanana, X. Guo, K.E. Avis, et al., J. Parenter. Sci. Technol. 47 (1), 22–5 (1993). 6. I. Markovic, Am. Pharm. Rev. 9 (6), 20-27 (2006). 7. D.R. Jenke, PDA J. Pharm. Sci. Technol. 59 (4), 265– 281 (2005) 8. FDA Container and Closure Integrity Testing in Lieu of Sterility Testing as a Component of the Stability Protocol for Sterile Products, (Rockville, MD, Feb. 2008). 9. L.S. Burrell, M.W. Carver, G.E. DeMuth, and W.J. Lambert, PDA J, Pharm. Sci. Technol. 54 (6), 449–55 (2000). 10. E. Kirsch et al., PDA J, Pharm. Sci. Technol. 51 (5), 195–202 (1997). 11. P. Kiang et al., J. Parenter. Sci. Technol. 46 (S2) (1992). ◆ BioPharm International www.biopharminternational.com December 2012 JANUARY 28th - 30th 2013 Sheraton San Diego Hotel & Marina San Diego, California, USA www.biomansummit.com North America’s Leading Biomanufacturing Event Uncovers Tomorrow’s Strategies to Help You Streamline Processes and Reduce Costs. Topic and Speaker Highlights Include: Developing a World-Class Quality Monitoring System Ronald C. Branning, Chief Quality Officer Genzyme USA Generating Competitive Advantage and Improved Performance through Lean Biomanufacturing Programming Divakar Ramakrishnan, VP, Manufacturing Elanco Animal Health Eli Lilly and Co. USA Analyzing the Risk of Supply Chain Sourcing and Manufacturing in Emerging Markets Andy Skibo, EVP Operations, MedImmune Facilities of the Future - What is the Future of Biomanufacturing? Jim Miller, VP and Head, Global Biologic Drug Substance Manufacturing - Genentech USA Understanding the Challenges Facing US Competiveness and Innovation in Bio Industries Charles Laranjeira, SVP, Technical Operations Cubist Pharmaceuticals Inc. USA Register now at www.biomansummit.com/1995 and pay only $1,995*USD *Please note that in order to book as a VIP delegate at the discounted price of $1,995 (+10% service fee) your company must be qualified as an end-user. WTG reserves the right to reject any registrations that do not fulfill this strict criterion. This offer is not available to existing Biomanufacturing Summit 2013 attending delegates, consultants or suppliers. For more information or to confirm your participation, please contact Pauline Duhart, Marketing Manager at pauline.duhart@wtgevents.com or phone 1 416 214 1144 Researched and Produced by: Operations Survey Survey: Optimizing Global Biopharmaceutical Operations Through Risk Mitigation and Management Phil Kaminsky, Jiyang Liu, and Julia Olsen-Claire A UC Berkeley survey provides insight into biopharma’s risk concerns and strategies. I ncreasing global competition and heightened c ustomer e x pec tations have for many years now encouraged enterprises in a variet y of industr ies to foc us on, and invest in, effective operations management. In most industries, managers no longer have to be convinced of the value of taking an integrated view of the design, analysis, and operation of their manufacturing, service, and logistics operations. Indeed, in many industries, operational excellence and a sophisticated approach to supplychain risk management based on flexibility, efficiency, and advanced tools for logistics network optimization are no longer a competitive advantage— they are necessary to compete. BACKGROUND Phil Kaminsky, Jiyang Liu, and Julia Olsen-Claire work at the CELDi Biopharmaceutical Operations Initiative at the University of California, Berkeley. 38 Consider, for example, the semiconductor industry. Thirty years ago, the semiconductor industry was a growing technology-focused industry and, for the first time, was beginning to face cost pressures. For years, the indust r y had foc used on super ior technolog y, and manufacturing was an afterthought. As long as the products were manufactured as they were envisioned by their inventors, there was little need to pay attention to capacity utilization, operational efficiency, inventory levels, or risk management; if you made them, profits would come. Howeve r, a s t he se m iconduc tor indust r y mat ured a nd compet it ive pressures grew, firms began to focus on operat ions, ut i l i z i ng resou rces BioPharm International www.biopharminternational.com December 2012 effectively and efficiently, optimizing systems, a nd perhaps most importantly, dealing effectively with u nce r t a i nt y a nd r i sk . Sig n i f ic a nt advances in the science of operations were required to bring about these changes, and by working collaboratively among themselves as well as with academia through organizations such as SEMATECH, semiconductor firms were able to make great strides, pushing the state-of-the-art in semiconductor operations to new heights. Biopharmaceutical firms now find themselves in a similar position. As the biopharmaceutical industry enters its fourth decade, it is entering a new, more mature phase. Revenue is growing, innovative business models and partnerships are being implemented, and products are coming to market. At the same time, this new maturity brings “adult-sized responsibility” (1). Although the science of biotechnology is advancing rapidly, with the promise of making an enormous impact on soc iet y, t he operat ions, supply chain, and log istics of biotechnology is not keeping pace. The ability of the industry to reach its potential requires systems that can produce and deliver products safely, reliably, and cost effectively to patients, while also allowing biopharmaceutical firms to successfully navigate the many risks inherent in the industry. It is becoming apparent that the biopharmaceutical supply chain presents a unique set of operational challenges—demand is highly uncertain and dependent on Operations Survey the results of clinical trials and competitors’ actions; supply is highly uncertain; biological processes are complex and incompletely understood; reg ulator y demands are significant and vary from region to region, including the existence of agencies in multiple jurisdictions that add layers of complexit y; contamination is difficult to detect and can have a significant impact; product failures can cost lives; IP concerns are significant; and capacity is extremely expensive and requires long lead times to build or acquire. ALL FIGURES ARE COURTESY OF THE AUTHORS RISK SURVEY Adding to the complexity, mechanisms for drug production are typically not standardized even within firms, technologies cont i nue to c ha nge, a nd gener ic drugs are poised to dramatically impact the industry (2). In recent years, this changing dy na mic has been a key focus of research at the University of California, Berkeley, culminating with the recent establishment of the CELDi Biopha r maceut ica l Operat ions Initiative (BOI), focusing on the deve lopme nt of c ut t i ng- e dge tools, techniques, and approaches to improve production systems, logistics systems, supply chain, inventory, and distribution within biopharmaceutical firms—essentially, biopharmaceutical operat ions. T he resea rc h i n it iat ive is jointly sponsored by member firms and the National Science F o u nd at io n ( N S F ) u nd e r t he Industry/University Cooperative Research Program. Key to the development of this initiative was a series of industry–academia workshops held at Berkeley (one sponsored by the NSF) in which the challenges and opportunities in biopharmaceutical operations management were explored. Across the industry, managers have Figure 1: Respondents indicate their level of concern about key risk categories, where the scale ranges from 1 to 5, with 1 meaning not concerned, 3 meaning concerned, 5 meaning extremely concerned. Raw materials supply risks Manufacturing supply reliability Contamination Outsourcing-related risk Forecast errors Catastrophic natural events Other 0 0.5 1 1.5 2 2.5 3 3.5 Figure 2: Respondents, as a percentage, indidcate how they make decisions regarding inventory level and/or the number of suppliers used (multiple selections were allowed). Network simulation tools Inventory/supply chain optimization tools Subject matter experts meeting Ad-hoc (no system looking specifically at this) Other (please specify) 0 an overlapping set of concerns, and are eager for better approaches, tools, and techniques that account for the unique characteristics of biopharmaceutical operations and help deal with these concerns. A common theme emerged from these workshops: the need for more effective risk-management tools and approaches. Many firms are specifically focusing on identifying and 20% 40% 60% hedging risks associated with their operations, and are eager to collaborate to improve tools, techniques, and approaches to do so. As a precursor to a concerted research effort in this area, the BOI surveyed nearly 300 industry members to explore attitudes about risk related to suppliers, raw materials, contamination, outsourcing, disposable technology, demand forecasting, inventory, December 2012 www.biopharminternational.com BioPharm International 39 Operations Survey Figure 3: Respondents indicate their level of concern about certain manufacturingrelated risks, where the scale ranges from 1 to 5, with 1 meaning not concerned, 3 meaning concerned, 5 meaning extremely concerned. Contamination Yield variability Increasing titers Disposables Human resources and training Other 0 1 2 3 4 Figure 4: Respondents rank how frequent key risks become an issue. 45.4% 10.1% 42% Every few years Once or twice a year About once a month More than once a month 40.3% lighted their concern that firms in the industry lack a “global” or “system” view of risks faced, and rather than developing a cohesive strateg y to minimize risk, consider risks one at a time, thereby ignoring their interactions. Surprisingly given these concerns, however, there is relatively little focus in the industry on detailed analysis of relevant data, relatively little formal quantification of risk, little formal modeling and simulation of risk or risk mitigation strategies, little focus on inventory optimization, and little measurement of uncertainty. In contrast, in many industries risk mitigation involves spending considerable resources collecting and analyzing data, assessing the types of variability in the data, optimizing resource utilization to mitigate risk, and developing rigorous models to understand where inventory and other buffers can most effectively be util i z e d t o h e d ge a g a i n s t r i s k . Surprisingly, for such a sophisticated industry in so many ways (e.g., compared with basic cons u me r pro duc t s or i ndu st r ia l equipment manufacturers), this industry has a qualitative view of risk management. This finding is particularly surprising given the vast amount of data that is collected in the industry. WHO TOOK THE SURVEY? and distribution, with a particular focusing on understanding which concerns are most significant, how firms measure these risks, and what mitigation strategies they currently have in place (see sidebar, “Survey Respondents”). Below is a summary of the survey’s key observations. SURVEY RESULTS Overall, firms are most concerned w ith qualit y risks, contamina- 40 tion risks, and risks associated with lack of visibility into contract manufacturing operations. More broad ly, f i r m s a re concerned with a broad spectrum of risk-related issues, including supplier risk, manufacturing reliability, inventory risks, cold-chain issues, a nd forecast ing-related risks. In preliminary interviews t hat accompa n ied t he su r vey, several respondents even high- BioPharm International www.biopharminternational.com December 2012 The BOI survey was distributed using a ma iling list compiled at industry meetings and from Berkeley’s BOI industry partners. Although the number of usable responses varies from question to que st ion, s u r vey s we r e at least partially completed. Fortysix percent of respondents work at biotech/ biopha r m f i r ms, and an additional 24% work at pharmaceutical firms. The bulk of t he rema ining respondents ident i f y t hemselves as equ ip - Operations Survey me nt or r aw-mate r ia l s uppl iers, or as employees of contract organizations. Twent y-five percent of t he respondents work at large companies (more than 50,000 employees), 20% work at very small firms (less than 100 employees), and the rest are dist r ib ute d s ome whe r e b e t we e n those two extremes. Slightly less than one quarter of respondents’ firms have no products on the market, while slightly less than half have 20 or more products on the market. In terms of annual s a le s, re sp onde nt s a l s o come from a broad spectrum of companies—35% are from large companies with $5 billion or more in annual sales, and the rest of t he respondents a re relat ively equally distributed over the range from no annual sales to $5 billion in annual sales. Respondents’ firms operate globally, with sales and manufacturing distributed around the world, and R&D most often concentrated on the US East and West Coasts, and in Western Europe. About 30% of survey respondents work i n R& D, a nd 2 0 % work in manufacturing; the rest are scat tered throughout var ious roles in engineering, quality, planning, and so for th. About half are managers or directors; the rest are either consultants, or in various roles ranging from t he C - su ite to i ndep e nde nt contractors. Note that the BOI team made no attempt to limit responses from a particular firm, so t hat t he resu lts ref lec t t he opinions of multiple respondents within a firm. OVERALL VIEW OF RISK O vera ll, respondents a re most concer ned w it h t he r isk s su rrounding manufacturing reliability and production cont a m i nat ion, fol lowe d by rawmaterial supply and outsourcing Figure 5: Respondents indicate their level of concern about certain contractmanufacturing risks, where the scale ranges from 1 to 5, with 1 meaning not concerned, 3 meaning concerned, and 5 meaning extremely concerned. On-time delivery/ Reliability Supplier outages Visibility into the process Quality issues Ensuring compliance with regulatory agencies Other 0 risk. Forecast errors, catastrophic natural events, regulatory risk, a nd I P t he f t r a n ke d f u r t he r behind, although all are of concern to respondents (see Figure 1). Interestingly, given risk concer ns and the inherent uncertainty in this industry, relatively few respondents rely on quantitative modeling tools to assess risk: 72% or respondents firms u s e fa i lu r e mo de a nd e f fe c t s a n a ly s i s ( F M E A) of r i sk , b ut fewe r t h a n 3 0 % u s e d i s c r e te event modeling, and fewer than 27% use Monte– Carlo analysis, tools that are standard in other industries. Indeed, while 70% of respondents monitor batches lost and on-time performance, half or fewer mon itor sa fet y stock levels or customer-service level, although these are key performance indicators that are useful for measuring the cost and effectiveness of risk mitigation strategies. Raw-material risk Of all of the raw-material related risks, single-sourcing stands as the most significant, followed by 1 2 3 4 contamination (other concerns included availabilit y, comparabi l it y ac ross vendors, e xt rac tables and leachables, and price f luctuations). These results are in spite of the fact that that only 37% of respondents indicate that their firms use multiple suppliers or similar strategies to mitigate t he r i sk , a lt houg h s ome responding firms are beginning to identify diversified sourcing as a key goa l i n t he produc tdevelopment stage. In addition to dual sourcing, firms turn to vendor audits, quality management systems, and large safety stock s of mater ia l i n order to mitigate these risks. One responde nt t a l ke d of aba ndon i ng a “just-i n-t i me” st rateg y due to the associated risk. Not surprisingly, inventory costs and inventor y levels are the key metrics t racked to assess raw-mater ia l acquisition performance (by 58% and 54% of respondents, respectively). Surprisingly, fewer than 15% of respondents e xpl ic it ly mention track ing qualit y metrics in this context. In addition, about ha lf of t he respondents December 2012 www.biopharminternational.com BioPharm International 41 Operations Survey Figure 6: Respondents indicate how well they explicitly model demand variability in their planning processes. 44.9% 18.0% 73.0% We don’t We model some upsides and downsides We use distributions to model percentiles of likelihood More than once a month conce r n s, re sp onde nt s rep or t primarily turning to high-temperature/short-time (HTST) pasteurization, physical segregation, and better assay technologies at roughly equivalent levels (each by between 45% and 47% of respondents). About 60% report experiencing a contamination event once or more a year, and roughly 14% experience such an event once a month or more frequently (see Figure 4). 29.8% ment ion foc using on t rack ing t he f rac t ion of single-sou rced raw materials. When determining the appropriate inventor ymanagement policies, a surprisingly large fraction of respondents (40%) of respondents do not use quantitative or scientific approaches. Slightly over half rely on optimization tools to make these decisions, and about 15% (possibly overlapping with the optimization users) also use simulation tools (see Figure 2). Manufacturing-related risk The most significant manufacturing-related risk as identified by respondents was contamination, followed in decreasing order by human resources issues, and y ield va r iabi l it y. Respondents were also concerned with increasing titers, but seemed less concer ned w it h t he c ha l lenges posed by disposables (see section below). Respondents also identified a variety of other concerns surrounding product validation, equipment reliability, and managing capacit y. Sec uring sufficient supply and was also noted a mong t he respondents’ comments (see Figure 3). Fou r pr i ma r y st rateg ies a re 42 used to mitigate risks including training, statistical analysis, quality management techniques, and a focus on improving both internal and external communication. About 40% of respondents identify training and education as a key ma nufac t ur ing r isk-reduction tool, while more than 30% identify statistical analysis and quality management. Moreover, re sp onde nt s rep e ate d ly h ig hlight the positive impact of good communications both internally and with CMOs and suppliers on manufacturing yields. When addressed specifically, ma nu fac t u r ing contaminat ion was reported to be a primary concern of 64% of the respondents (see next section). Contamination events Of the various issues and concer ns sur rounding contamination, media contamination is by far the largest concern: 39% of respondents ident if ied t his as their most significant contamination-related concern. Crossbatc h cont a m i nat ion wa s a distant second at 19% (followed by lack of detectability of cont a m i n at ion, a nd e x t r ac t able s and leachables). To address these BioPharm International www.biopharminternational.com December 2012 Contract manufacturing A l m o s t 6 0 % o f r e s p o n d e nt s report using CMOs for some or all of their manufacturing. Although respondents show concern about all aspects of their relationships with CMOs (e.g., reliability, visibility, flexibility, and IP) the most dominant concerns are around quality and compliance (these are the only categories for which a plurality of respondents indicated “extreme concern”). To address these risks, the most commonly noted approaches include a comprehensive focus on prescreening and monitoring CMOs, frequent audits, and keeping a representative on the CMO site (and duals ou r c i n g whe r e p o s sible a nd reasonable) (see Figure 5). Disposables Over half of respondents (53%) currently use disposable technology for commercial manufacturing, and a total of 83% either plan to or are likely to use disposable commercial manufacturing technology in the future. The most reported significant risks around this technology focus, in decreasing order of reported significance are: comparabilit y as products are scaled up for commercial runs, timely availability of supplies from suppliers, and extractables and leachables. To add ress t hese r isk s, respondents turn to many of the same Operations Survey approac hes t hat t hey u se for ma nag i ng raw-mate r ia l r i sk s, includ ing using mu lt iple sup pliers where possible, stringent vendor qualification and audits, extensive qualit y control testing and validation, and carefully constructed contracts specifying minimum inventory levels. Forecasting and demand variability About half (49%) of the respondents consider forecast errors a significant risk; less than a quarter (24%) are not concerned. In spite of this, 30% do not explicit ly mo de l de ma nd va r i abi lit y in their planning process, and another 45% only model some upside and downside scenarios; only 18% explicitly model demand distributions (see Figure 6). By far, the dominant approach (70% of the respondents) to dealing with demand uncertainty is to hold extra inventory—most of the other cited approaches have to do with securing extra capacity, either internally or through relationships with CMOs. About one third of respondents explicitly track forecast accuracy by comparing point forecasts with actual demand, and about one quarter suggest that their firms do not explicitly track forecast accuracy. Distribution Of the various risks associated with finished-goods distribution, cold storage and expiration-related issues are more of a concern than contamination, theft, and counterfeiting. No single approach stands out in the reported mitigation methods for these concerns, although respondents seem to continually explore alternative suppliers and methods for packaging, shipping, and storage (in particular, they mention continual quality auditing of transportation service providers, secure packag- Survey respondents t t t t t "ENJOJTUFSFEPOMJOFVTJOH4VSWFZ.POLFZJOUIFGJSTURVBSUFSPG SFTQPOEFOUTJODMVEJOH o #JPQIBSNB o 1IBSNB o $.0$30TVQQMJFSTFUD 'JSNTBMFTUPUBMFE o CJMMJPO o oNJMMJPO 'JSNFNQMPZNFOUUPUBMFE o FNQMPZFFT o FNQMPZFFT .BOVGBDUVSJOH3%BOE4BMFTUPPLQMBDFJOUIF64$BOBEB-BUJO"NFSJDB 8FTUFSO&VSPQF"GSJDB.JEEMF&BTU"TJB"VTUSBMJB/FX;FBMBOE t 'VODUJPOBMBSFBTJODMVEFE o 3% o .BOVGBDUVSJOH o 0UIFSMJTUFEBSFBTJODMVEFETVQQMZDIBJOBOERVBMJUZ GJOBODFQMBOOJOHMPHJTUJDTBOEFOHJOFFSJOH t 5JUMFTJODMVEFE o 7JDFQSFTJEFOUTFOJPSWJDFQSFTJEFOU$MFWFM o %JSFDUPS o .BOBHFS o 0UIFSUJUMFTMFTTUIBO JODMVEFETVQFSWJTPSBOEDPOTVMUBOU ing, tracking systems, diversification, inventor y management software, and so forth). More than 80% of responding firms store inventory in multiple sites, selecting these sites in most cases for their proximity to manufacturing sites or their low operating costs. Once finished goods shift to distributor control, only 25% of respondents are not concerned with distributors’ risks, although only 20% are confident in their visibility into distributors’ risks, suggesting a key area of potential improvement. CONCLUSION AND NEXT STEPS This sur vey prov ides init ia l i nsights i nto t he c u r rent state of risk management and r i s k- m it i g at io n s t r at e g i e s i n t he biopha r maceut ica l i ndus try. In subsequent surveys, the BOI tea m i ntend s to e x plore s p e c i f ic r i s k- m it i g at ion ap p roaches and metrics c urrently employed by progressive firms, in order to develop a better understanding of which metrics, tools, and approaches are particularly useful. At the same time, there is clearly a need within the biopharmaceutical industry for analytical tools and approaches that account for the specific characteristics of this industry, and that use the vast quantities of data available to help managers make more rigorous, informed, model-based decisions to manage and mitigate the complex set of risks faced by the industry. REFERENCES 1. Ernst & Young, “Beyond Borders: Global Biotechnology Report,” 2007. 2. R. Johnson and P. Kaminsky, “Biopharmaceutical Operations: Developing the Science,” PharmaFocus Asia 9 (2008). z December 2012 www.biopharminternational.com BioPharm International 43 CORPORATE CAPABILITIES www.biopharminternational.com SGS Life Science Services Company Description SGS Life Science Services is a leading contract service organization providing clinical research services, analytical development, biologics characterization a n d q u a l it y c o nt r o l testing. SGS provides clinical trial management (Pha se I to I V ) a nd ser vices encompassing bioa na ly t ic a l te st i ng , data management, biostatistics, and regulatory consultancy. SGS also offers contract analytical services (detailed below) that include analytical chemistry, microbiology, stability studies, method development, and protein analysis. With the recent acquisition of Vitrology, SGS expands its service offering for biologics in the areas of cell bank and virus seeds characterization, raw material and bulk harvest testing (sterility, mycoplasma, viruses), final product testing for residual DNA, and safety consultancy services. SGS is the world’s leading inspection, verification, testing, and certification company. SGS LIFE SCIENCE SERVICES 75 Passaic Ave. Fairfield, NJ 07004 TELEPHONE 866.747.5003 FAX 973.244.1823 EMAIL lss.info@sgs.com WEBSITE www.sgs.com/lifescience NUMBER OF EMPLOYEES Life Science Services worldwide: 1500 SGS Group: 70,000 YEAR FOUNDED 1878 44 Technical Services t Quality control testing of raw materials, APIs, and finished products t Monograph testing (USP, EP, BP, and JP) t A nalytical method development and validation t Antibody product analysis t Bioana lysis, mass spectrometr y and immunoassays t Biologics safety testing (endotoxin, virus and mycoplasma) t Cell line characterization t Container testing (extractables and leachables) t Environmental Monitoring t Glycosylation Analysis BioPharm International December 2012 t t t t t t t t Host cell impurity testing Virus testing (characterization of Cell Banks) Molecular biology services Medical device testing Microbiological testing Protein/peptide analysis and quantification Safety testing (in vitro) Stabilit y testing according to ICH guidelines or customer specifications t Water system validation t Clinical trials from Phase I-IV t Data management and statistics Facilities With truly global coverage and a strong local footprint, in North America, SGS has facilities in Lincolnshire (Illinois), Fa irf ield (New Jersey), West Chester (Pennsylvania), and Mississauga (Canada), as well as a clinical trial management office in Gaithersburg (Maryland). SGS’s laboratories operate according to high quality standards (cGMP, GLP, ISO 17025) and have been inspected by FDA or local regulator y authorities. Markets Served SGS ser ves various life science companies, including pharmaceutica l, biopharmaceutical, biotechnolog y, and medical device manufacturers. SGS operates a global network of 19 Life Science Services laboratories with facilities in the US, UK, C a nad a, Belg iu m, Fra nc e, Germa ny, India, China, Taiwan, Singapore, and Switzerland. The Top 20 pharmaceutical and biopharmaceutical companies trust SGS as a partner for their quality control testing needs. ADVERTORIAL THE LARGEST NETWORK OF CONTRACT ANALYTICAL LABS SGS LIFE SCIENCE SERVICES SGS Life Science Services is a leading contract service organization providing analytical development, biologics characterization, biosafety testing, quality control product release, as well as Phase I-IV clinical research services. Whether your organization is large and global or small and regional, rely on SGS as your partner for outsourced testing. Operating a harmonized network of 19 laboratories in 11 countries, SGS offers lean quality standards, reliability and regulatory / technical expertise... all within close proximity to you. © 2012 SGS Société Générale de Surveillance SA – All rights reserved CONTACT US: Europe: +32 10 42 11 11 North America : +1 866 747 5003 Asia : +65 63790 111 pharmaqc@sgs.com SGS IS THE WORLD’S LEADING INSPECTION, VERIFICATION, TESTING AND CERTIFICATION COMPANY WWW.SGS.COM/PHARMAQC WHEN YOU NEED TO BE SURE Global Markets Navigating Emerging Markets In an introduction to a new BioPharm International series on manufacturing within global markets, the author provides key considerations for getting started. Jill E. Sackman, DVM, PhD, is a senior consultant at Numerof & Associates, Inc. (NAI), St. Louis, MO, www.nai-consulting.com. 46 T he global market for pharmaceuticals continues to grow rapidly, and in response, manufacturers are moving to align their market strategy and product pipeline to meet emerging, unmet needs. Companies exploring opportunities in global markets face dynamic demographic and disease trends, changing market demands, and evolving regulatory requirements—all of which differ from one country to another. This complex environment makes planning for global market entry into both developed and developing countries a moving target. In the face of such complexity, organizations need to be prepared to navigate constantly changing, inconsistent, and diverging regulatory and market trends in a structured, but flexible manner that minimizes rework and maximizes value. Whether the goal is to outsource the manufacture of US products to India to lower costs, or to market imported or domestically developed products in China, a consistent approach for understanding market trends and navigating foreign regulation of pharmaceuticals is crucial for developing a successful global market strategy. With that BioPharm International www.biopharminternational.com December 2012 Chad Baker/Getty Images Jill E. Sackman said, it is important to allow for variability in regional requirements (sometimes even within a single country), market demands, and product pipelines. Over the next year, I’ll be tackling some of the region-specific challenges that pharmaceutical companies face in a new BioPharm International column. This introductory article provides some key considerations for navigating and managing diverse market and regulatory trends across the globe in a consistent manner. DEVELOP A COMMON APPROACH TO NAVIGATING REQUIREMENTS The globalization of pharmaceutical markets has fueled an emphasis on improving the regulation of these industries across developed and developing countries. Ideally, steps towards harmonization would be among the goals of improving regulation. Countries are instead taking independent strides at varying paces to improve the regulation of markets that are very different from each other. Across the board, submission requirements vary and enforcement resources are limited, which can contribute to frustrating waiting periods and prolonged product-approval timelines. Global Markets This degree of regulatory variability may seem daunting, but approaching regional requirements in a consistent manner can make the process more manageable. Although regulatory oversight and enforcement varies from country to country, there are common regulatory steps that play an integral role in shaping each stage of the product development, approval, manufacturing, and marketing lifecycle. Regardless of the region or product in question, the regulatory process will likely involve the following: product classification, preclinical testing, clinical evaluation, product registration, manufacturing/production approval, quality system management, and compliance with good manufacturing practice (GMP) as well as good clinical practice (GCP), and good laboratory practice (GLP). At each step in the regulatory process, authorities may establish approval requirements, regionally accepted best practices, recognized credentials and certifications, and/ or quality system requirements. The controls that are put in place at each step in the regulatory process can—and will—differ from country to country, and from product to product. Being able to identify and understand these controls for any given country or region will be crucial because they may have implications for timelines, processes, cost, and revenues. Moreover, understanding the nature and stringency of these regulatory measures will be important. These factors may impact how effectively a product can be brought to market in other regions, under a completely different set of regulatory requirements. evolved over time may help organizations prepare to navigate trends in a rapidly changing region. As an example, China and Brazil are developing health technology infrastructures along the lines of the UK’s National Institute for Health and Clinical Excellence (NICE). Understanding what that process has looked like could be helpful in preparing for future regulatory demands in these developing markets. DETERMINE THE MOST EFFICIENT ALLOCATION OF RESOURCES As part of developing a consistent approach, it will be important to determine the most efficient way to identify and collect information and to manage your organization’s ongoing presence in foreign markets. Consider centralizing the allocation of resources, as appropriate, to minimize duplication of effort to understand regulatory and market trends for a particular region. By centralizing these efforts, you can ensure that one division of your organization is not building networks or partnerships in regions where you already have those relationships developed. To ensure that your company is able to navigate foreign market trends and changing regulatory requirements on Continued on p. 57 GAIN INSIGHT INTO REGIONAL MARKET TRENDS The implications of population growth, demographic changes, and shifting lifestyles on disease trends and market demands differ tremendously from region to region. As part of determining market potential within a given region, it will be important to understand these trends within the context of your technical requirements (e.g., product specifications, materials.) and operational needs. Additionally, regional health policy and reimbursement trends will have a tremendous impact on successful market entry, and should serve as criteria for developing your market strategy. Specifically, it will be important to understand in-region: access to healthcare, epidemiological trends, current clinical treatment paradigms (care paths), market demand (i.e., taking into consideration the patient population as well as clinical insight from in-region providers), competitive set/competing products, reimbursement for similar products , and data requirements (i.e., clinical and health economics) for market access. It is also important that the decisions your organization makes as part of its global strategy account not only for current trends, but also for the expected or anticipated future environment. Looking at how similar markets have December 2012 www.biopharminternational.com BioPharm International 47 Boot Camp: Tech Guide Glycan Analysis: A Primer I n this fourth part of a series of primers with training experts from the National Institute for Bioprocessing Research and Training (NIBRT), Pauline Rudd, PhD, professor of glycobiology at University College Dublin (UCD), discusses glycan analysis. NIBRT provides training, educational, and research solutions for the international bioprocessing industry in state-of-theart facilities. Located in South Dublin, it is based on an innovative collaboration between UCD, Trinity College Dublin, Dublin City University, and the Institute of Technology Sligo. KEY DEVELOPMENT AND MANUFACTURING CONSIDERATIONS BioPharm: Can you provide a brief overview of what exactly glycan analysis targets and its importance in bioprocessing? Rudd: Most glycoproteins, and almost all of the new biological drugs, are proteins with sugars attached to them. These sugars are important for the safety and efficacy of drugs, so it is necessary to be able to control the processing of the sugar structures to make sure that the drug is as effective as possible. There are many aspects of developing and processing a drug that require having analytical technologies for glycosylation. For example, in the beginning of a drug-development process, one needs to understand the role of the sugars on the protein being used. On erythropoietin, for example, the sugars must be multiantennary and fully capped with sialic acid; otherwise, the erythropoietin will only be in the patient for a few minutes, whereas if it is completely sialylated, it will be present in the patient for 3 hours or more, during which time it will be able to be effective in stimulating the production of red blood cells. It is key to monitor the production process to make sure that the erythropoietin has the sugar structures that provide the full benefits of glycosylation. It is always important for biologics developers to understand exactly how the sugars in their product are going to modulate the functions of the drug in 48 BioPharm International www.biopharminternational.com the patient. Once this knowledge is obained, drug developers can define an optimal glycan profile. The next stage of bioprocessing, clonal selection, requires identifying a high-producing clone that has the ability to fully glycosylate the molecules with the optimal sugars. When selecting clones, glycan analysis enables the producer to determine the complement of glycoenzymes that are operating within a particular clone. The next aspect of glycosylation that must be checked is the potential introduction of an antigenic epitope. If one is making a product in a nonhuman cell line, such as mouse cells, it is necessary to check the levels of alpha-galactose and N-glycolyl-neuraminic acid residues, which may be antigenic to humans. After the candidate clones are selected, the process of producing the protein from the cells must be monitored. Taking samples during the process allows one to assess whether the media composition is optimal for producing the desired glycosylation profile. In process development, optimizing the media is necessary to produce high levels of correctly folded proteins as well as the desired post-translational modifications, including glycosylation. BioPharm: What role do glycans play in the manufacturing stage? Rudd: When making a biologic product, one needs to track the glycans, which will help to determine the best time to harvest the product. When one reaches the downstream processing stage, high-performance liquid chromatography (HPLC) is often used to select subfractions of the product. Being able to analyze the glycans to ensure that their subfractions are correctly glycosylated is crucial. This information will be used during conversations with regulatory authorities so that the agency can agree on specifications for the drug product. Glycan analysis will be part of that specification because it will be part of the regulators assessment of the drug’s safety and efficacy. It will also be necessary to demonstrate to regulators that the process is robust and December 2012 Sveta Demidoff/Getty Images NIBRT’s Pauline Rudd on what to expect when performing glycan analysis. Leveraging the Unique Attributes of Chromatography Resins to Drive Productivity and Decrease Cost of Goods 0N-DEMAND WEBCAST Register free atwww.biopharminternational.com/resins EVENT OVERVIEW In order to address current industry manufacturing challenges and demands, process development scientists must balance a number of factors when designing a downstream process including capacity, resolution, process flexibility, disposability, throughput and cost. Increased titers and product demand have caused a substantial bottleneck in downstream processing for both antibodies as well as recombinant proteins. There is a greater need to purify molecules more efficiently and to reduce the costs associated with purification. This webcast will discuss New solutions on the chromatography side of purification to improve process performance and increase process flexibility How high performance chromatography can impact biotherapeutic cost of goods Examples of how to generate more productive and cost effective processes to address downstream challenges Who Should Attend Biopharmaceutical companies, biotech companies, and contract manufacturing organizations with titles that include: Process Development Process Engineering Quality Assurance/Control/Validation Key Learning Objectives Discuss new approaches for the downstream purification processes to obtain improved impurity clearance and maximize yields Discuss how to obtain improved purity and throughput, while increasing process flexibility within a purification scheme Discuss how chromatography can benefit downstream processing and impact cost of goods and overall productivity Speakers Shelly Cote Parra Sr. Applications Scientist Life Technologies Christine Gebski Head of the POROS® Resin Business and Global Applications Life Technologies Moderator Angie Drakulich Editorial Director BioPharm International Anything to do with manufacturing Presented by Or, any customer interested in the manufacturing/production of biomolecules such as monoclonal antibodies, therapeutic proteins, and vaccines. Sponsored by For questions, contact Sara Barschdorf at sbarschdorf@advanstar.com Boot Camp: Tech Guide can be reproduced for batch and lot release. Then comes the consideration of long-term storage. Although in general, glycans are quite stable, it is important to ensure that glycans do not change by testing after degradation and stability studies. Functional assays are also important to determine whether a product, IgG, for example, is able to bind to the desired receptor and not to those receptors that can cause side effects in the patient. Some of these questions require an understanding of how glycosylation modulates activity of the drug. Overall, there are many reasons to perform glycan analysis and many ways to approach it. It is therefore important to understand the question being asked before deciding which method to use. COMMON CHALLENGES IN CONTROL AND TECHNIQUE BioPharm: W hat common challenges is the industry facing today when performing glycan analysis? Rudd: Glycan processing is difficult to control because it involves a complex process that involves the expression of genes (the genes carry the code for the glycosylating enzymes) as well as the delivery of monosaccharides on nucleotide donors to grow the glycans. There are many things to control, and nearly 600 proteins are required to build the glycans as well as to transport the glycoproteins into the correct organelle for complete glycan processing. If one is over-expressing a protein, it is possible to exhaust some of the glycosylation machinery. It is not uncommon to find incomplete structures attached to a protein, which indicate that the processes to build the sugars has not operated on all copies of the glycoprotein going through the secretory pathway. Understanding how to get the cells to express at a level where the rest of the machinery can cope is just one challenge. 50 BioPharm International Another difficulty is determining what to do when the glycosylation is going wrong. One needs to understand in depth how media components and metal ions, for example, can alter glycan analysis. The natural cell is exquisitely tuned and responds very finely to its environment. It is difficult to replicate this robustness in a bioprocesssor. BioPharm: You mentioned that industry uses various techniques for glycan analysis. What are the most commonly used instruments and techniques? Rudd: Glycan analysis depends on a series of separations technologies that exploit different physical properties of the oligosaccharides. There are several ways to approach it. If you are focused on analyzing released glycans, you need to have an optimized method for releasing them. There are many glycoconjugates, but from the point of view of the pharmaceutical companies the most commons ones are N-linked glycans. There is an enzyme called PNGase F that can be used to remove them from proteins of most species. After that stage, various separations technologies can be used to separate glycans according to mass/ charge, charge, size, and lipophilicity by techniques such as hydrophilic interaction chromatography (HILIC), ion-exchange chromatography, reverse-phase chromatography, or capillary electrophoresis. BioPharm: What gaps still exist in glycan-related technology and instrumentation? Rudd: NIBRT uses a lot of special instrumentation, but we are working to get the analytical technologies miniaturized and as straightforward as we can. We have a robot that can accommodate 96 well plates, for example, so that one can put the samples on the instrument and come back later to put the released sugars on a HPLC machine or use mass spectrometry for analysis. We also have the capability to run lin- www.biopharminternational.com December 2012 ear samples, meaning that we can take samples one at a time from a bioprocesser every few hours. Going forward, industry will be looking to miniaturization, automation, and, particularly, automated data analysis. REGULATORY ISSUES BioPharm: What regulatory expecta- tions exist for glycan analysis when developing a biologic compared with a legacy product? Rudd: Actually, there is a debate at the moment. Regulators need companies to report critical features of the glycosylation, but in some cases, it is not clear what “critical” features include. There is a need for more basic research to clarify these questions. For some molecules, such as IgG, it is known that the Fc glycosylation modulates effector function, so the regulators can ask for a full glycan analysis of IgG. One can report the sialylated structures, the levels of galactosylated, fucosylated, and bisected structures, because it is known that each of these features can modulate a function. To get the information, teams need to perform a complete analysis of IgG and present the data in a way that answers the questions about critical features that affect the safety and efficacy of their product. If one is working with erythropoietin, it is necessary to report the percentages of different antennary structures as well as the extent of sialylation, because this critically affects the pharmacokinetcs of the drug. One needs to report levels of N-glycol-neuraminic acid, alpha(1,3) linked galactose as well as levels of xylose and alpha(1-3)-linked fucose, because these are potential antigenic epitopes. In general, regulatory expectations are getting higher because the technolog y is getting better. In the past, none of us really understood the implications of glycosylation in therapeutics Continued on p. 56 MAY 20-22, 2013 Sheraton San Diego Hotel and Marina San Diego Call For Papers Contributed Papers Submission Deadline: February 13, 2013 SUBMISSION DETAILS: www.aaps.org/nationalbiotech Bioanalytical Best Practices Bioanalytical Methods for Sample Cleanup Preparation of biological samples for chromatographic analyses. 52 BioPharm International www.biopharminternational.com The choice of sample preparation method should depend on the quality of the data required. tion method should depend on the quality of the data required. It makes little sense to invest weeks of development time attempting to achieve pg/mL sensitivity for a screening assay. However, it may be important to invest such time to develop and validate a method for a lead drug candidate undergoing human safety assessments that are subject to FDA regulatory scrutiny. Indeed, validating analytical procedures is the process of determining a suitable method that is capable of providing useful analytical data. It is important to bear in mind that a method that is valid in one situation could be invalid in another (10, 11). In the pharmaceutical industry, the most common biological sample matrix is plasma. Moreover, it is common practice to dilute troublesome matrices like urine or cerebrospinal fluid with plasma and apply previously developed plasma extraction protocols. Drugs are most commonly isolated from plasma using one (or occasionally, a combination) of either liquid–liquid extraction, protein precipitation, or solid phase extraction. Other less common choices include column-switching (LC–LC), affinity extraction, and ultrafiltration. LIQUID-LIQUID EXTRACTION Liquid-liquid extraction (LLE) using organic solvents offers sample cleanup with analyte December 2012 Image: PASIEKA/Science Photo Library/Getty. H igh-throughput bioanalytical methods are essential to support the rapid discovery and development of drugs in the pharmaceutical industry. Liquid chromatography coupled with tandem mass spectrometric detection (LC–MS/MS) is considered as the benchmark analytical methodology for quantifying new chemical entities in biological fluids (1–4). Because of the high sensitivity and selectivity of LC–MS/MS, the time required for method development and subsequent sample analysis is dramatically reduced. Rigorous chromatographic resolution of analytes and/ or tedious sample extraction protocols are typically not required even when complex biological matrices are used. Most chromatographic techniques have matured and automation is now commonplace (5–7). Nevertheless, with common sample analysis times of less than three minutes, the bottleneck in sample analysis has become the sample preparation step. Sample preparation is still considered to be a slow and labor-intensive process, and it is rare for an analyst to be able to inject samples directly into an LC–MS/MS system with no pretreatment. The importance of sample preparation stems from three major concerns—removing interferences from the biological sample matrices, concentrating the analyte(s) of interest, and improving analytical system performance (8). An industry survey noted a marked increase in methods requiring limits of quantitation of less than 1 ppb, and the trend toward trace analyses is not diminishing (9). Roger N. Hayes is vice-president and Optimized sample preparation techgeneral manager of laboratory sciences niques that provide high enrichat MPI Research, ment factors become crucial for 54943 North Main Street, these dilute concentrations. Mattawan, MI 49071. The choice of sample prepara- Bioanalytical Best Practices enrichment steps, and is a rugged off-line sample preparation process that is well suited for routine high-t hroughput LC – M S/ MS analysis. The basic concept of LLE is to partition an analyte into a volatile organic solvent away f rom polar proteins and lipids that remain in an aqueous phase. The organic phase is removed, evaporated, a nd t he sample reconstituted for injection onto an LC –MS/MS system. By careful choice of organic solvent, LLE is amenable to automation in a 96-well format (12–15). In order, the acceptability of solvents for automated LLE is methyl t-butyl ether > 95/5 hexane/ethanol >> ethyl acetate. LLE does, on occasion, suffer from emulsion formation, which may be resolved by extended centrifugation. LLE can also be performed in the solid state by using diatomaceous earth (Hydromatrix or Celite). Several such products are available commercially for performing supported liquid extractions (SLE) in a 96-well plate format. PROTEIN PRECIPITATION Plasma sample preparat ion by protein precipitation (PPT) is the most widely used technique for LC–MS/MS analysis because of its simplicity, low cost, and universality. PPT is amenable to automation in a 96-well format (16). Precipitation of plasma proteins is most commonly performed by using organic solvents like acetonitrile or methanol. Following denaturation, the sample is cent r i f uge d a nd t he sup er nat a nt is d i rec t ly i njec ted. However, organic solvents are inefficient in precipitating proteins and often require signif icant dilution of the plasma sample, typically by two- to three-fold. Overcoming the impact of dilution by injecting larger volumes of the plasma extract may be precluded when using reversed-phase HPLC because of the high organic content. Evaporation of the extracted samples to near dryness followed by reconstitution in an appropriate solvent is usually required. In order, the preference of solvent for automated PPT is 95/5 (v/v) acetonitrile/acetone > acetonitrile >> methanol. ability, ultimately such a loss in sensitivity limits the achievable limit of quantification. 96 -well filter plates are available commercially and they provide the analyst the ability to conduct, in an automated fashion, the protein crash procedure, the mix step, and the filtration of protein precipitants in the same 96-well flow-through plate. Assessing the potential for ion suppression over the elution time is an important step in developing a rugged method. SOLID-PHASE EXTRACTION Alternative choices for precipitation of plasma proteins include trichloroacetic acid (TCA) or zinc sulfate. Reagents like TCA and zinc sulfate, however, are unable to remove small proteins, polypeptides, and salts, thereby contributing to high ionic strength of the supernatant that can subsequently suppress ionization and attenuate LC–MS/MS response. Despite the attractiveness of PPT as a rapid approach to sample preparation, potential drawbacks exist. For example, because only proteins are removed, ion s u p p r e s s io n f r o m c o - e lut i n g matrix components can significantly reduce sensitivity, especially when using electrospray ionization. Therefore, assessing the potential for ion suppression over the elution time is an important step in developing a rugged method. Although using stable isotope-labeled internal standards can compensate for response vari- December 2012 The third alternative to sample cleanup is solid-phase extraction (SPE). SPE met hods consist of loading samples onto pre-conditioned sorbent-filled cartridges, often arranged in a 96-well plate format. Loaded samples are then washed with an appropriate solvent, and the analyte(s) is subsequently eluted. Method selection and extract cleanliness are determined by the retention mechanism and the ability of the wash stage to effectively remove endogenous components. For example, ionic interactions, as represented by cation and anion exchange, offer a greater degree of analyte selectivity and, therefore, extract cleanliness when compared to C18-based sorbents. In general, extracts from SPE are cleaner than those from PPT. SPE ga i ne d i n p opu la rity because of its compatibility with automation, especially with sorb e nt mate r ia l pac ke d i nto a 96 -well format plate (17–18). Te c h nolo g ic a l i mp r o v e m e nt s include the development of polymeric SPE sorbents that no longer suffer from sorbent drying problems while enjoy ing extended working pH ranges. Taking advantage of the full pH range of the sorbent, a specific pH and organic modulated SPE (i.e., an optimized SPE) method can be developed to provide clean sample extracts. T he hyd roph i l ic-hyd rophobic nature of these polymeric sup- www.biopharminternational.com BioPharm International 53 Bioanalytical Best Practices p or t s i s a me nable to ge ne r ic extraction techniques. A ge ne r ic p r o to c ol s ho u ld achieve high recovery; however, high recoveries do not necessarily correlate with high sensitivity in LC–MS/MS. Achieving high sensitivity is usually a trade-off between recovery and chemical interference or ion suppression. Nevertheless, high sensitivity and high recovery are achievable by selective retention of a basic drug using strong cation exchange SPE. Fortunately, the majority of drug candidates have a basic functionality that can be leveraged for selective retention on an appropriate strong cation exchange SPE material. A s de s c r ib e d, SPE met ho d s often involve evaporation and subsequent reconstitution of the eluent before LC–MS/MS analysis. T hese steps not only take time and effort, but can also lead to the loss of valuable sample. Therefore, the ability to elute in ver y small volumes of solvent is desirable to minimize sample pr e pa r at ion t i me a nd r e duc e sample loss. Low sorbent mass and novel 96-well plate designs, including SPE pipette tips and discs, have allev iated some of these concerns (19). Another approach to consider is the direct coupling of SPE to the LC–MS/MS system (i.e., online SPE) (20). Differences in flow rates du r ing load a nd elut ion steps afford additional opportunities to enhance the extraction process. For example, sufficiently high flow rates can induce turbulent flow chromatography that actually involves a combination of size-exclusion and adsorption phenomena (21–24). If the analyte fraction has a high enough affinity for the stationary phase inside the pores, then it w ill remain there until a solvent w ith the 54 BioPharm International appropriate strength desorbs it. Online SPE methods have the potential to significantly enhance sensitivity because no dilution of sample occurs. Eliminating analyte collection, evaporation, reconstitution, and injection not only improves reproducibility, but also saves time, labor, and solvents. CONCLUSION T he genera l idea of sa mple cleanup is that all elements of a method should contribute to its required sensitivity and selectivity. Issues to consider when selecting a bioanalytical method for sample cleanup should include what matrix the analy te is in, the detection limit and dynamic range required, the number of samples to be analyzed, analyte stability to extraction, and the amount of matrix available. The final method should be orthogonal to maximize selectivity and reduce ion suppression. If C18 SPE is used for sample extract ion, t hen t he a na lyst should c on sid e r c at ion e xc h a n ge or phe nyl colu m n c h romatog ra phy. Alternatively, if strong cation exchange SPE was selected for sample extraction, then any reverse-phase LC method can be used. The sample cleanup method should be assessed for recovery, selectivity, precision, accuracy, and ruggedness. Formal validation may also be required (25–27). REFERENCES 1. T.R. Covey, E.D. Lee, and J. Henion, Anal. Chem. 58 (12) 2453–2460 (1986). 2. H. Fouda et al., J. Am. Soc. Mass Spectrom. 2, 164–167 (1991). 3. E.C. Huang et al., Anal. Chem. 62, 713–725 (1990). 4. R.S. Plumb et al., Xenobiotica, 31 (8–9), 599–617 (2001). 5. D. Mole, R.J. Mason, and R.D. McDowall, J. Pharm. Biomed. Anal. 11 (3), 183–190 (1993). www.biopharminternational.com December 2012 6. E. Doyle et al., Anal. Proc. 26, 294– 295 (1989). 7. M. Jemal, Biomed. Chromatogr. 14 (6), 422–429 (2000). 8. R.D. McDowall et al., J. Pharm. Biomed. Anal. 7 (9), 1087–1096 (1989). 9. R.E. Majors, LCGC North America, 20, 1098–1113 (2002). 10. B.A. Persson, J. Vessman, and R.D. McDowall, LC-GC Int. 11, 160–164 (1998). 11. B.A. Persson, J. Vessman, and R.D. McDowall, LC-GC Int. 10 (9), 574–576 (1998). 12. N. Zhang, K.L. Hoffman, W. Li, and D.T. Rossi, J. Pharm. Biomed. Anal. 22, 131–138 (2000). 13. Z. Shen, S. Wang, and R. Bakhtiar, Rapid Commun. Mass Spectrom. 16 (5), 332–338 (2002). 14. J. Zweigenbaum et al., Anal. Chem. 71 (13), 2294–2300 (1999). 15. M. Jemal, D. Teitz, Z. Ouyang, and S. Khan, J. Chromatogr. B 732 (2), 501– 508 (1999). 16. D. O’Connor, D.E. Clarke, D. Morrison, and A.P. Watt, Rapid Commun. Mass Spectrom., 16 (11), 1065–1071(2002). 17. C. Sottani, C. Minoia, M. D’Incalci, M. Paganini, and M. Zucchetti, Rapid Commun. Mass Spectrom. 12 (5) 251– 255 (1998). 18. H. Simpson et al., Rapid Commun. Mass Spectrom., 12 (2) 75–82 (1998). 19. R.E. Majors. New Designs and Formats in Solid-Phase Extraction sample preparation, LCGC Europe 12 (1) 2–6 (2001). 20. F. Beaudry, J.C. Yves Le Blanc, M. Coutu, and N.K. Brown, Rapid Commun. Mass Spectrom 12 (17) 1216–1222 (1998). 21. H.M. Quinn and J.J. Takarewski, “Improvement in Chemical Analyses” International Patent Number WO97/16724 (May, 1997). 22. M. Jemal, Zh. Ouyang, B.C. Chen, and D. Teitz, Rapid Commun. Mass Spectrom. 13 (11) 1003–1015 (1999). 23. J.T. Wu, H. Zeng, M. Qian, B.L. Brogdon, and S.E. Unger, Anal. Chem. 72 (1) 61–67 (2000). 24. M. Jemal, Y. Qing, and D.B. Whigan, Rapid Commun. Mass Spectrom. 12 (19) 1389–1399 (1998). 25. J.R. Kagel, W. Donati, L.E. Elvebak, and J.A. Jersey, Amer. Lab., 33 (24) 20–23 (2001). 26. V.P. Shah et al., Eur. J. Drug Metab. 16 (4) 249–255 (1991). 27. A.R. Buick, et al., J. Pharm. Biomed. Anal. 8 (8–12) 629–637 (1990). ◆ Final Word Patents and Postapproval Batch Testing Can postapproval FDA filings immunize pharma companies from patent lawsuits? It shall not be an act of infringement to make, use, offer to sell, or sell . . . a patented invention . . . solely for uses reasonably related to the development and submission of information under a Federal law which regulates the manufacture, use, or sale of drugs or veterinary biologic products (2). But what happens if the company is sued for infringement after it obtains approval from FDA? Until recently, most in the legal industry would have said that postapproval batch testing is not protected by Hatch–Waxman. As recent as 2011, in Classen Immunotherapies, Inc. v. Biogen IDEC, the Federal Circuit Court of Appeals—the final authority on most patent matters—decided that the safe harbor does not apply to “information that may be routinely reported to the FDA, long after marketing approval has been obtained” (2). Classen had accused Biogen and GlaxoSmithK line (GSK) of infringing Classen’s patent relating to an immunization method (3). The patented method involved screening immunization schedules and selecting and administering the schedule that presented the lowest risk of developing certain immune-mediated chronic disorders later in life (4). As part of an FDA study, Biogen and GSK used the patented methods to provide vaccines, advise on immunization schedules, and report adverse vaccine effects to FDA (5). Biogen and GSK contended that If the Hatch–Waxman Act protects postapproval batch testing, the value of patents on analytical methods, particularly covering biosimilars, may significantly decrease. their activity fell within the safe harbor (6). But the Federal Circuit disagreed, concluding that the provision only provides a safe harbor to expedite the development of information for regulatory approval of generic counterparts of patented products. GSK and Biogen were held not immune from Classen’s suit. But i n Aug u st 2 012 , t he s a me cou r t decided in Momenta Pharmaceuticals, Inc. v. Amphastar Pharmaceuticals, Inc., (7) that the safe harbor provision indeed might immunize post-approval activities. The patent at issue in Momenta covered a method for analyzing batches of enoxaparin, a synthetic version of the blood-thinning agent heparin (8). Because of enoxaparin’s unique chemical makeup, FDA requires batch analysis as a condition for the post-FDA approval sale of the drug (9). Momenta claimed that Amphastar’s quality control testing of its enoxoparin batches i n f r i nged Moment a’s patent; A mphast a r argued that its testing fell within Hatch– Waxman’s safe harbor because FDA required the testing (10). The Federal Circuit determined that § 271(e)(1) unambiguously covers any activity reasonably related to developing M. Freeman/PhotoLink/Getty Images F DA requires that pharmaceutical companies create and maintain pre-approval batch records for both generic and brand drugs. In general, a company can do so without risking infringing any patents covering manufacture of a drug. Congress enacted the so-called “safe harbor” provision of the Hatch– Waxman Act in 1984 specifically allowing pharmaceutical companies to test their products prior to obtaining regulatory approval to market them (1). Under that provision: Kevin Murphy is a partner with the New York office of Frommer Lawrence and Haug LLP. Andrew Nason is an associate in the DC office of the same firm, KMurphy@ flhlaw.com. The views expressed in this article are solely those of the authors and are not to be attributed to Frommer Lawrence and Haug LLP or any of its clients. December 2012 www.biopharminternational.com BioPharm International 55 Final Word and submitting information under a federal law that regulates the manufacture, use, or sale of drugs regardless of whether the activity occurs pre- or postapproval (11). The decision has major consequences for the industry. If the Hatch–Waxman Act protects postapproval batch testing, the value of patents on analytical methods, particularly covering biosimilars, may significantly decrease. C h ie f Judge R ade r, who aut hor e d t he m ajo r it y o p i n ion in Classen, issued a lengthy and shar ply worded dissent in Mo m e n t a , c r it ic i z i n g h i s f e l low judges for failing to follow Classen (12). Judge Moore, who authored the majority opinion in Momenta and a dissent in Classen, saw it differently. She squared the Classen and Momenta decisions not i ng t hat, i n Classe n, there was no requirement that Biogen or GSK submit any data to FDA. By contrast in Momenta, FDA required analytical records for each batch of enoxoparin produced in order to maintain regulatory approval (13). W het he r one c a n re conc i le these two decisions, the Supreme Cour t could ver y well end up resolv ing t he issue. GSK f iled a petition for certiorari asking t he Supreme C ou r t to c la r i f y “[w]hether the Federal Circuit’s i nte r p r e t at io n o f § 271(e)(1) [in Classen], which a rbit ra r ily restricts the safe harbor to preapproval activities, is faithful to statutory text that contains no such limitation, and decisions of this Court rejecting similar ef for ts to impose extratext ual limitations on the statute” (14). The Supreme Court may welcome the opport unit y to prov ide its guidance on the scope of Hatch– Waxman’s safe harbor—an issue t he h igh cou r t has add ressed twice before (15)— especially in light of this seeming inconsistenc y in Federa l Circ u it precedent in t his a rea of t he law. REFERENCES 1. Eli Lilly & Co. v. Medtronic, Inc., 496 U.S. 661, 669-71 (1990). 2. 35 U.S.C. § 271(e)(1) (2012). 3. 659 F.3d 1057, 1060, 1070 (Fed. Cir. 2011). 4. Ibid. at 1060. 5. Ibid. at 1070-72. 6. Ibid. at 1070. 7. 686 F.3d 1348 (Fed. Cir. 2012). 8. Ibid. at 1351. 9. Ibid. at 1353. 10. Ibid. at 1352-53. 11. Ibid. at 1354-55. 12. Ibid. at 1361-76. 13. Ibid. at 1357-58. 14. Petition for a Writ of Certiorari at i., GlaxoSmithKline v. Classen Immunotherapies, Inc., No. 11-1078 (U.S. Feb. 28, 2012). 15. Merck KGaA v. Integra Lifesciences I, Ltd., 545 U.S. 193, 206-07 (2005); Eli Lilly & Co. v. Medtronic, Inc., 496 U.S. 661 (1990). ◆ Boot Camp: Tech Guide Continued from p. 50 and regulatory requirements were to define the glycosylation as well as one could. Our understanding has moved a long way since then, and regulations are far more demanding. Readers can listen to the full interview with Dr. Rudd via an audio podcast on BioPharmInternational.com. ◆ KEY TAKEAWAYS t 6OEFSTUBOEUIFSPMFPG TVHBSTPOZPVSQSPUFJO t #FBXBSFPGWBSJPVT JOTUSVNFOUBUJPOPQUJPOT UPQFSGPSNTFQBSBUJPO BOBMZTFT t ,FFQVQUPEBUFXJUI UIFJNQSPWFNFOUTJO CJPJOGPSNBUJDTGPSEBUB JOUFSQSFUBUJPO 56 BIOPHARM’S BOOT CAMP GUIDES Below is a list of all the BioPharm International Boot Camp guides that exist to date, including articles and podcast interviews. Check them out on www.BioPharmInternational.com/BasicTraining. If there is a topic we haven’t covered yet that you would like to see in a future issue, please email the editor at aritter@advanstar.com. Business guide topics: t Outsourcing Strategies t Considerations when Working with Suppliers t GMP Facilities and Operations Management t The Drug-Development Game: Risks and Rewards t Product Differentiation t Virtual Biotech Setups t Industry Training t Commercializing a Compound t Project Management t Pipeline Growth t Implementing Knowledge Management t Changing Your Business Model Technical guide topics: t Biosimilars Development t Reference-Product Dilemmas t Downstream Processing t Upstream Processing t Product Characterization t Aseptic Processing and Fill/Finish BioPharm International www.biopharminternational.com December 2012 Ad Index Company Navigating Emerging Markets – Page AAPS National Biotech 51 Althea Technologies Inc 16, 17 Catalent Pharma Solutions 60 EMD Millipore 5 Eppendorf North America 2 IBC Life Sciences 29 Eurofins Lancaster Laboratories 30, 31 Life Technologies 21, 49, Gatefold Nova Biomedical 58, 59 Parker Hannifin Domnick Hunter 27 Patheon Pharmaceutical Svc Inc 9 Rentschler Biotechnologie GmbH 19 SGS Life Sciences 44, 45 Woodley Equipment Co 47 WTG-10th Annual Biological Pro 37 Wyatt Technology Corp 13 an ongoing basis, organizations also frequently decide to pursue partnerships with in-region resources that are familiar with current and impending clinical, regulatory, reimbursement, and competitive landscapes. Additionally, in-region resources are prepared to handle cultural nuances that you may not anticipate, which may ultimately have an impact on timelines, costs, and successful market entry. Knowing when to outsource is important, but managing in-region partnerships closely is equally crucial. Most regions are moving to regulate in-region firms as an extension of the manufacturer that partnered with them, and therefore, centralized management of partners is more crucial now than ever before. A consistent mechanism for ensuring that you have the necessary auditing capabilities in place will be key to the success of these relationships. CELL LINES Continued from p. 47 BE PREPARED FOR CHANGE The demands placed on drug-product companies operating on a global basis are changing dramatically. New regulatory requirements have a significant impact on matters, such as product design, commercialization, and market-entry strategies. Additionally, demographic and disease trends, evolving policy changes and shifting market demands complicate navigating foreign markets in an efficient manner. These dynamic, unpredictable, and region-specific considerations introduce a great deal of risk for companies. However, for companies that have developed the capabilities to anticipate and adapt to change, the current and impending global environment presents tremendous opportunity. ◆ You can hear additional tips from Dr. Sackman in a podcast interview on www.BioPharmInternational.com. FROM Rapid Development of HIGH PERFORMANCE CHO Based Cell Lines ELEMENTS OF BIOPHARMACEUTICAL PRODUCTION SERIES, FOURTH EDITION Newly Updated Led by Dr. Anurag Rathore. For anyone involved in or planning to start process development, characterization and/or validation activities. www.selexis.com LAB EQUIPMENT $ 129.95 Airflow Diagnostic Tools save on these and other educational resources at www.industrymatter.com | 800.598.6008 CORPORATE CAPABILITIES www.biopharminternational.com Nova Biomedical NOVA BIOMEDICAL 200 Prospect Street Waltham, MA 02454 TELEPHONE 781.894.0800 Company Description Nova Biomedical is a technology company based in Waltham, Massachusetts that provides stateof-the-art automated cell culture analyzers for the biopharmaceutical market. In 1998, Nova introduced the first line of chemistry analyzers for the biotechnology industry—the BioProfile® series. In 2007, Nova added the BioProfile® Flex series with an expanded menu and realtime analysis of key metabolites, nutrients, and gases in cell culture media. These fully automated, multi-test systems simultaneously measure the following parameters: pH, glucose, lactate, glutamine, glutamate, pO2, pCO2, ammonium, sodium, potassium, phosphate, IgG, calcium, chloride, and cell viability and density. In 2011, we announced the BioProfile CDV Automated Cell Density/viability analyzer that can measure up to 80 million cells/mL and provide results in less than 3 minutes. BioProfile analyzers are intended to optimize process development, enhance process reliability and reproducibility, improve manufacturing yield and quality, and reduce testing time and cost. In fact, BioProfile analyzers can replace up to seven different instruments and measurement techniques while dramatically decreasing space requirements and capital equipment, labor, and reagent costs. An optional On-Line Autosampler allows the user to connect multiple spinner flasks, bioreactors, and other vessels directly to the BioProfile analyzer, and establish user-defined sampling frequencies and feedback instructions for each reactor vessel. All results are stored in a Data Management System for subsequent retrieval or transmission to a data historian. key nutrients, metabolites, and gases in cell culture and fermentation media. Our all-in-one BioProfile FLEX analyzer provides immediate measurement of key chemistries, cell density/ viability, IgG, phosphate, and osmolality in cell culture media to provide a total picture of cell growth in a single instrument. The BioProfile FLEX OPC Connectivity is a complete plug and play solution that facilitates connectivity with all OPC-compliant systems. Our optional On-Line Autosampler connects up to 10 bioreactors to one BioProfile FLEX, providing real-time testing. BioProfile Analyzers feature: t Simultaneous measurement of up to 15 key parameters t A full 15 test profile including IgG in eight minutes t Automatic one button operation t Direct sampling from syringes, tubes, flasks, and pipettes t 0.5 mL sample size t No sample dilution or centrifugation t Automatic analyzer calibration Cell Density/Viability Analyzer Nova’s BioProfile CDV incorporates advanced technology for rapid, accurate, high resolution measurement of cell density and cell viability at a moderate cost. Based on the widely recognized trypan blue exclusion method, BioProfile CDV combines advanced technology in robotics, optics and computer algorithms in a highly automated analyzer. t Easy to use, intuitive interface t 12-position sample tray for walk-away sample analysis t High resolution vision system t Advanced computer algorithms for fast image processing and classification BioProfile products provide assays necessary to address the testing requirements of process development, pilot plant, and production facilities. Developed in conjunction with leading biotechnology companies, these test menus enable control of the most critical constituents of cell culture media. FAX 781.894.5915 EMAIL info@novabio.com WEBSITE www.novabiomedical.com 58 Major Products Cell Culture Chemistry Analyzers BioProfile® series automated chemistry analyzers are designed for real-time, rapid analysis of BioPharm International December 2012 ADVERTORIAL One Automated Analyzer for Fast Comprehensive Cell Culture Analysis The Power of One Flexible Modular Design BioProfile FLEX can reduce cell culture analysis time, labor, and operating costs by consolidating multiple analyzers into a single, easy to use workstation. BioProfile FLEX can be customized with 1–4 modules to consolidate up to 15 vital cell culture tests: Module 1: Glucose, Lactate, Glutamine, Glutamate, Ammonium, pH, PCO2, PO2, Sodium, Potassium, Calcium Module 2: Cell Density/Cell Viability Module 3: Osmolality Module 4: IgG One compact workstation, up to 15 Cell Culture assays, including IgG One small, 1 mL sample conserves cell culture mass and end product One fast, 6 minute analysis saves at least one hour over multiple instrument analyses One integrated data source simplifies data collection, analysis, and archiving One validation saves time vs validation of multiple instruments New IgG Module A new IgG measurement combines protein binding methodology with photometric endpoint detection, to provide: IgG results in less that 6 minutes No sample preparation or centrifugation 0.1 to 10.0 g/L measurement range Other Options On-Line Autosampler with automated sampling from up to 10 bioreactors OPC Connectivity automates data and control commands Test Menu Gluc Lac Gln Glu NH4+ pH PO2 PCO2 Na+ K+ ZZZ%LR3URÀOH)/(;FRP Ca++ CD CV Osm IgG