58
The Science & Business of Biopharmaceuticals INTERNATIONAL INTERNATIONAL BioPharm International NOVEMBER 2015 Process Chromatography I Cell Culture I Viral Clearance Technology Volume 28 Number 11 November 2015 Volume 28 Number 11 STERILITY ASSURANCE UPSTREAM PROCESSING IMPLICATIONS OF CELL CULTURE CONDITIONS ON PROTEIN GLYCOSYLATION PEER-REVIEWED ESTABLISHING PROCESS DESIGN SPACE FOR A CHROMATOGRAPHY PURIFICATION STEP SUPPLY CHAIN DIVERSIFYING THE GLOBAL HEPARIN SUPPLY CHAIN www.biopharminternational.com

STERILITY ASSURANCEimages2.advanstar.com/PixelMags/biopharm/pdf/2015-11.pdf · 756-5255 or email: [email protected]. UBM Life Sciences provides certain customer contact data (such

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

  • The Science & Business of Biopharmaceuticals

    INTERNATIONALINTERNATIONAL

    Bio

    Ph

    arm

    Intern

    atio

    nal

    NO

    VEM

    BER 2

    015

    Pro

    cess C

    hro

    mato

    gra

    ph

    y I C

    ell C

    ultu

    re I V

    iral C

    leara

    nce

    Tech

    no

    log

    y

    Vo

    lum

    e 2

    8 N

    um

    ber 1

    1

    November 2015

    Volume 28 Number 11

    STERILITY

    ASSURANCE

    UPSTREAM PROCESSING

    IMPLICATIONS OF CELL

    CULTURE CONDITIONS ON

    PROTEIN GLYCOSYLATION

    PEER-REVIEWED

    ESTABLISHING PROCESS DESIGN

    SPACE FOR A CHROMATOGRAPHY

    PURIFICATION STEP

    SUPPLY CHAIN

    DIVERSIFYING THE

    GLOBAL HEPARIN

    SUPPLY CHAIN

    www.biopharminternational.com

    ES701252_BP1115_cv1.pgs 11.05.2015 21:58 ADV blackyellowmagentacyan

  • PHARMACEUTICAL n HEALTH SCIENCES n FOOD n ENVIRONMENTAL n CHEMICAL MATERIALS

    ©2015 Waters Corporation. Waters, ACQUITY QDa and The Science of What’s Possible are registered trademarks of Waters Corporation.

    Gain confidence in glycan, peptide, and

    oligonucleotide analysis with mass detection.

    ES700041_BP1115_CV2_FP.pgs 11.04.2015 02:42 ADV blackyellowmagentacyan

  • INTERNATIONAL

    BioPharmThe Science & Business of Biopharmaceuticals

    EDITORIALEditorial Director Rita Peters [email protected] Editor Agnes Shanley [email protected] Editor Susan Haigney [email protected] Editor Randi Hernandez [email protected] Science Editor Adeline Siew, PhD [email protected] Director Dan Ward [email protected] Editors Jill Wechsler, Jim Miller, Eric Langer, Anurag Rathore, Jerold Martin, Simon Chalk, and Cynthia A. Challener, PhD Correspondent Sean Milmo (Europe, [email protected]) ADVERTISING

    Publisher Mike Tracey [email protected]/Mid-West Sales Manager Steve Hermer [email protected] Coast Sales Manager Scott Vail [email protected] Sales Manager Chris Lawson [email protected] Sales Manager Wayne Blow [email protected] List Rentals Tamara Phillips [email protected] 877-652-5295 ext. 121/ [email protected] Outside US, UK, direct dial: 281-419-5725. Ext. 121 PRODUCTION Production Manager Jesse Singer [email protected] AUDIENCE DEVELOPmENT Audience Development Rochelle Ballou [email protected]

    UBm LIfE SCIENCES

    Tom Ehardt, EVP & Senior Managing Director, Life Sciences Tom Mahon, Senior VP, Finance Georgiann DeCenzo, EVP & Managing Director, UBM Medica Mike Alic, EVP, Strategy & Business Development Dave Esola, VP & Managing Director, Pharm/Science Group Johanna Morse, VP & Managing Director, CBI/IVT Becky Turner Chapman, VP & Managing Director, Veterinary Group Joy Puzzo, VP, Marketing & Audience Development Francis Heid, VP, Media Operations Jamie Scott Durling, Director, Human Resources

    UBm AmERICAS

    Simon Foster, Chief Executive Officer Brian Field, Chief Operating Officer Michael Bernstein, Head of Legal

    UBm PLC

    Tim Cobbold, Chief Executive Officer Andrew Crow, Group Operations Director Marina Wyatt, Chief Financial Officer Dame Helen Alexander, Chairman

    © 2015 Advanstar Communications Inc. All rights reserved. No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical including by photocopy, recording, or information storage and retrieval without permission in writing from the publisher. Authorization to photocopy items for internal/educational or personal use, or the internal/educational or personal use of specific clients is granted by Advanstar Communications Inc. for libraries and other users registered with the Copyright Clearance Center, 222 Rosewood Dr. Danvers, MA 01923, 978-750-8400 fax 978-646-8700 or visit http://www.copyright.com online. For uses beyond those listed above, please direct your written request to Permission Dept. fax 440-756-5255 or email: [email protected].

    UBM Life Sciences 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 UBM Life Sciences 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 UBM Life Sciences’ 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.

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

    K. A. Ajit-Simh President, Shiba Associates

    Rory Budihandojo Director, Quality and EHS Audit

    Boehringer-Ingelheim

    Edward G. Calamai Managing Partner

    Pharmaceutical Manufacturing

    and Compliance Associates, LLC

    Suggy S. Chrai President and CEO

    The Chrai Associates

    Leonard J. Goren Global Leader, Human Identity

    Division, GE Healthcare

    Uwe Gottschalk Vice-President,

    Chief Technology Officer,

    Pharma/Biotech

    Lonza AG

    Fiona M. Greer Global Director,

    BioPharma Services Development

    SGS Life Science Services

    Rajesh K. Gupta Vaccinnologist and Microbiologist

    Jean F. Huxsoll Senior Director, Quality

    Product Supply Biotech

    Bayer Healthcare Pharmaceuticals

    Denny Kraichely Associate Director

    Johnson & Johnson

    Stephan O. Krause Director of QA Technology

    AstraZeneca Biologics

    Steven S. Kuwahara Principal Consultant

    GXP BioTechnology LLC

    Eric S. Langer President and Managing Partner

    BioPlan Associates, Inc.

    Howard L. Levine President

    BioProcess Technology Consultants

    Herb Lutz Principal Consulting Engineer

    Merck Millipore

    Jerold Martin Independent Consultant

    Hans-Peter Meyer Lecturer, University of Applied Sciences

    and Arts Western Switzerland,

    Institute of Life Technologies.

    K. John Morrow President, Newport Biotech

    David Radspinner Global Head of Sales—Bioproduction

    Thermo Fisher Scientific

    Tom Ransohoff Vice-President and Senior Consultant

    BioProcess Technology Consultants

    Anurag Rathore Biotech CMC Consultant

    Faculty Member, Indian Institute of

    Technology

    Susan J. Schniepp Fellow

    Regulatory Compliance Associates, Inc.

    Tim Schofield Senior Fellow

    MedImmune LLC

    Paula Shadle Principal Consultant,

    Shadle Consulting

    Alexander F. Sito President,

    BioValidation

    Michiel E. Ultee Principal

    Ulteemit BioConsulting

    Thomas J. Vanden Boom VP, Biosimilars Pharmaceutical Sciences

    Pfizer

    Krish Venkat Managing Partner

    Anven Research

    Steven Walfish Principal Scientific Liaison

    USP

    Gary Walsh Professor

    Department of Chemical and

    Environmental Sciences and Materials

    and Surface Science Institute

    University of Limerick, Ireland

    ES701449_BP1115_003.pgs 11.06.2015 01:03 ADV blackyellowmagentacyan

  • 4 BioPharm International www.biopharminternational.com November 2015

    Contents

    BioPharmINTERNATIONAL

    BioPharm International integrates the science and business of

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

    peer-reviewed technical solutions to enable biopharmaceutical professionals

    to perform their jobs more effectively.

    COLUMNS AND DEPARTMENTS

    BioPharm International ISSN 1542-166X (print); ISSN 1939-1862 (digital) is published monthly by UBM Life Sciences 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: IMEX Global Solutions, P. O. Box 25542, London, ON N6C 6B2, CANADA. Canadian GST number: R-124213133RT001. Printed in U.S.A.

    BioPharm International is selectively abstracted or indexed in: • Biological Sciences Database (Cambridge Scientif c Abstracts) • Biotechnology and Bioengineering Database (Cambridge Scientif c Abstracts) • Biotechnology Citation Index (ISI/Thomson Scientif c) • Chemical Abstracts (CAS) • Science Citation Index Expanded (ISI/Thomson Scientif c) • Web of Science (ISI/Thomson Scientif c)

    Cover: PLAINVIEW/Maria Toutoudaki/Getty Images; Dan Ward

    6 From the Editor Biopharma and contract providers must tread carefully amid changing market dynamics. Rita Peters

    8 Regulatory Beat New program emphasizes quality, risk, and global collaboration. Jill Wechsler

    10 Perspectives on Outsourcing Better process develop-ment is creating industry benchmarks for bioprocessing. Eric Langer

    48 Compliance Notes How to ensure archive records can be retrieved. Siegfried Schmitt

    49 Troubleshooting There are many factors to consider when choosing viral clearance methods.Cynthia A. Challener

    53 New Technology Showcase

    53 Ad Index

    54 Biologics News Pipeline

    FILL/FINISHBest Practices for Sterility Assurance in Fill/Finish OperationsRandi HernandezExperts discuss best practices to achieve acceptable sterility assurance levels for aseptically filled products. 14

    UPSTREAM PROCESSINGImplications of Cell Culture Conditions on Protein GlycosylationRichard Easton and Michiel E. UlteeThe authors present a review of the techniques commonly used for glycosylation analysis. 20

    DOWNSTREAM PROCESSINGThe Development ofProcess Chromatographyin BioprocessingSusan HaigneyIndustry experts discuss the development of process chromatography in bioprocessing. 26

    PEER-REVIEWEDEstablishing Process Design Space for a Chromatography Purification Step: Application of Quality-by-Design PrinciplesHui XiangThis case study reviews how quality-by-design principles can be implementedin an intermediate chromatography purification step that usescation-exchange chromatography. 28

    GLOBAL SUPPLY CHAINDiversifying the Global Heparin Supply Chain: Reintroduction of Bovine Heparin in the United States?David Keire, Barbara Mulloy, Christina Chase, Ali Al-Hakim, Damian Cairatti, Elaine Gray, John Hogwood, Tina Morris, Paulo A.S. Mourão, Monica da Luz Carvalho Soares, and Anita SzajekThe global supply chain for bovine and porcine heparin and regulatory considerations are examined. 36

    SUPPLY CHAIN

    Piloting Track-and-Trace ImplementationRobert CelesteVirtual pilot programs examine scenarios that may occur while implementing serialization requirements for theUS Drug Supply Chain Security Act. 43

    QUALITYInvestigating BiologicsSusan Schniepp and Andrew HarrisonThe authors discuss performing investigations of biological products. 46

    Volume 28 Number 11 November 2015

    fEATURES

    ON THE WEBwww.biopharminternational.com

    Future of Bioprocessing eBook

    BioPharm’s The Future of Bioprocessing eBook features articles on advanced biologics, single-uses systems, market demand, patent reviews, automation, and more!

    To read the eBook, visit:

    BioPharmInternational.com/FutureofBioprocessingeBook

    BioPharmINTERNATIONAL

    The Science & Business of Biopharmaceuticals

    THE FUTURE OF

    October 2015

    e B O O K S E R I E S

    ES701503_BP1115_004.pgs 11.06.2015 02:37 ADV blackyellowmagentacyan

  • 052.A1.0103.A © 2015 Eppendorf AG.

    www.eppendorf.com/cic • 800-645-3050

    ImagineThe new Eppendorf Cell Imaging Consumables

    > Innovative design for

    ergonomic handling

    > TC treatment for reliable

    cell attachment

    > Precise planarity for high

    resolution microscopy and

    autofocus imaging systems

    Whether you perform live or fixed

    cell imaging, seed cells on microscope

    slides or cover glasses, image a single

    cell or a full 96 well plate, the new

    Eppendorf Cell Imaging Consumables

    offer tailored solutions for the best

    optical performance in your cell

    imaging experiments.

    ES700029_BP1115_005_FP.pgs 11.04.2015 02:41 ADV blackyellowmagentacyan

  • 6 BioPharm International www.biopharminternational.com November 2015

    From the Editor

    Biopharma and

    contract providers

    must tread carefully

    amid changing

    market dynamics.

    Next Steps in Outsourcing Relationships

    In an industry where change is the norm, biopharma companies must learn

    to successfully navigate the financial, business, regulatory, and scientific

    ups and downs of the market. In the fourth part of the 2015 CPhI Annual

    Industry Report, Looking beyond the Global Pharma Horizon (1), industry repre-

    sentatives commented on dynamics in biologics development and the contract

    services market, and how challenges and strategic approaches in the two sectors

    may direct the industry moving forward.

    Increased funding in the emerging bio/pharma sector, changing customer

    attitudes and business practices, regulations, a global supply chain, niche

    technology offerings, and untapped markets will shape the contract services

    market, writes Gil Roth, president of the Pharma and Biopharma Outsourcing

    Association. Most critical, however, is the ways in which contract manufactur-

    ing organizations (CMOs) and contract development and manufacturing orga-

    nizations (CDMOs) learn from the industryÕs past.

    In ÒCMO/CDMO Challenges and Opportunities,Ó Roth notes that recent

    acquisitions in the contract services market were motivated by the desire to

    integrate service offerings, or acquire niche technologies to attract earlier phase

    clients with the goal of retaining that business through commercial manufac-

    turing. At the same time, the improving economy has enabled more capital

    investment in facilities at biomanufacturing firms, with a resulting shift of

    some operations in house. In addition, a focus on orphan drugs with smaller

    batch sizes may shift technology requirements and outsourcing relationships.

    Hedley Rees, managing consultant at PharmaFlow, highlights differences in

    the manufacture and supply of large-molecule biologic products versus small-

    molecule drugsÑpotential pitfalls in the drug development processÑin ÒWhat

    Does the Future Hold for Biopharmaceutical Outsourcing?Ó

    Rees cites the effects of even minor changes in the production process, chal-

    lenges in sourcing raw materials, analytical methods to detect changes during

    manufacture, product sensitivity to environmental factors, and the current

    model of pharmaceutical distribution as potential opportunities for failure. In

    addition, advanced therapy medicinal productsÑgene therapies, somatic cell

    therapies, and tissue-engineered productsÑwill demand closer ties between the

    manufacturer and healthcare system versus the one-size-fits-all batch process-

    ing traditionally used with current blockbuster therapies.

    In the present fee-for-service outsourcing model, projects are directed by a

    contract; changes must be negotiated, with both cost and time implications.

    The risk for the contract service provider is low, versus a risk- and-reward-

    sharing model.

    ÒThe banana skin waiting for the unsuspecting pharmaco is that this new era

    of biologics needs a different approach to outsourcing,Ó Rees warns.

    Contractors can offer technical expertise that biologics companies need;

    however, some biopharma companies are considering more in-house operations.

    A move away from outsourced operations may drive the contract service market

    to think more about a risk-sharing model.

    Rees identifies factors that will drive discussions between drug owners and

    contractors including the use of a quality-by-design approach; supply chain

    reporting and control; patenting of process knowledge; the cost in of commer-

    cialization; and the availability of qualified personnel.

    For both parties, a careful eye on market changes and development needs, as

    well as some strategic hand-holding, may avert some nasty slips or falls.

    Reference 1. CPhI, Annual Report 2015, Part IV, Looking beyond the Global Pharma Horizon, online

    www.cphi.com/europe/networking/cphi-pharma-insights, accessed Nov. 2, 2015. ◆

    Rita Peters is the editorial director of

    BioPharm International.

    ES699965_BP1115_006.pgs 11.04.2015 01:08 ADV blackyellowmagentacyan

  • Be a superhero. You can with our unmatched

    chromatography portfolio.

    Learn more about our new

    Chromabolt¨ prepacked columns at:

    Process scale chromatography can be challenging Ð

    even for a superhero. But with the broadest portfolio

    of chromatography resins, columns, and unparalleled

    Whether youÕre tackling higher and higher titers, racing

    the clock, or conquering bottlenecks, we can help

    you be the superhero.

    Affinity. Ion Exchange. Reversed Phase. Expertise.

    Your cape is waiting at

    EMD Millipore is a division of Merck KGaA, Darmstadt, Germany

    EMD Millipore, the M logo and Chromabolt are registered trademarks of Merck KGaA, Darmstadt, Germany. © 2015 EMD Millipore Corporation, Billerica, MA USA. All rights reserved. PS-SBU-13-09113. 2/2015

    www.emdmillipore.com/Chromabolt

    www.emdmillipore.com/chromatography

    support from EMD Millipore, you can make it happen.

    ES700058_BP1115_A7_FP.pgs 11.04.2015 02:42 ADV blackyellowmagentacyan

  • Be a superhero.

    You can with our unmatched

    chromatography portfolio.

    Learn more about our new

    Chromabolt® prepacked columns at:

    Process scale chromatography can be challenging –

    even for a superhero. But with the broadest portfolio

    of chromatography resins, columns, and unparalleled

    Whether you’re tackling higher and higher titers, racing

    the clock, or conquering bottlenecks, we can help

    you be the superhero.

    Affinity. Ion Exchange. Reversed Phase. Expertise.

    Your cape is waiting at

    Merck Millipore, the M logo and Chromabolt are registered trademarks of Merck KGaA, Darmstadt, Germany. © 2015 EMD Millipore Corporation, Billerica, MA USA. All rights reserved. PS-SBU-13-09113. 2/2015

    www.merckmillipore.com/Chromabolt

    www.merckmillipore.com/chromatography

    support from Merck Millipore, you can make it happen.

    ES700040_BP1115_B7_FP.pgs 11.04.2015 02:42 ADV blackyellowmagentacyan

  • 8 BioPharm International www.biopharminternational.com November 2015

    Regulatory Beat

    Vis

    ion

    so

    fAm

    eri

    ca

    /Jo

    e S

    oh

    m/G

    ett

    y Im

    ag

    es

    After two years of planning and analy-

    sis, FDA officials are moving forward

    with implementation of the Program

    Alignment plan to better coordinate agency

    field inspections with product reviews from

    FDA research centers. The aim is to reduce

    redundant processes and to provide more exper-

    tise in evaluating today’s more complex and

    varied production systems for drugs and bio-

    logics. The growing number of pharmaceutical

    ingredients and finished products imported

    from abroad, moreover, heightens the need for

    risk-based oversight and increased collaboration

    with foreign regulatory counterparts to avoid

    duplicate inspections.

    Transforming oraThe reorganization of FDA’s 5000-person field

    force represents the most important change

    since the Office of Regulatory Affairs (ORA)

    was formed, says Melinda Plaisier, ORA chief

    and associate commissioner for regulatory

    affairs. This Program Alignment initiative,

    announced in September 2013 and further clar-

    ified in February 2015 (1), is dissolving ORA’s

    five regional offices and establishing

    commodity-based and vertically inte-

    grated inspection programs for drugs,

    biologics, medical devices, tobacco

    products, food, and bioresearch mon-

    itoring that will operate out of ORA’s

    20 district offices.

    For drugs, Pla isier expla ined

    at the PDA/FDA Joint Regulatory

    Conference in Washington, DC in

    September 2015, Alonza Cruse will be

    director for Pharmaceutical Quality

    Operations, which will have a cadre

    of pharmaceutical inspectors divided

    into four management teams. Anne

    Reid is acting director for Biological

    Operations, with two management teams, and

    Jan Welch heads up three teams for medical

    devices. Some product team directors also will

    head district offices.

    These teams of specialized investigators will

    gain greater technical expertise through train-

    ing, which should help them keep pace with

    manufacturing changes and new technology,

    especially those inspectors with sub-specialties

    in, for example, sterile drugs, compounding,

    APIs, or  combination products. Pharmaceutical

    inspectorate members also will be part of

    the Center for Drug Evaluation and Research

    (CDER) product review teams so that they will

    fully understand development and manufactur-

    ing issues involved in a new therapy and can

    produce pre-approval inspection reports that

    reflect a common understanding of pertinent

    production and quality concerns.

    Plaisier emphasized that the ORA overhaul

    is a “work in progress,” and that many final

    decisions and individual assignments are still

    to come. Questions remain about the num-

    ber of field management teams for each pro-

    gram, where these will be located, and how to

    align some 2000 investigators into the different

    review programs, she explained. These transi-

    tion activities will continue through the com-

    ing year, with the goal of starting up the new

    FDA Overhauls Inspection OperationsNew program emphasizes quality, risk, and global collaboration.

    Jill Wechsler is BioPharm

    International’s Washington editor,

    Chevy Chase, mD, 301.656.4634,

    [email protected].

    fDa also seeks to halt violative

    imports more quickly by

    de-linking import alerts

    from warning letters.

    ES699852_BP1115_008.pgs 11.04.2015 00:12 ADV blackyellowmagentacyan

  • November 2015 www.biopharminternational.com BioPharm International 9

    regulatory Beat

    model in fiscal year 2017. ORA is

    looking to develop metrics to mea-

    sure the impact of these changes

    internally, along with enhanced

    training programs, new work plan-

    ning systems, and more central-

    ized laboratory operations.

    CoorDinaTing ComplianCeClear, coherent enforcement

    strategies with reduced layers of

    review involve closer collabora-

    tion between Center staff and field

    inspectors to eliminate duplicate

    case workups and to speed inspec-

    tion findings to manufacturers,

    explained Tom Cosgrove, direc-

    tor of the Office of Manufacturing

    Quality (OMQ) in the CDER Office

    of Compliance (OC). These changes

    should accelerate re-reviews of

    plants looking to regain compli-

    ance status and “not leave firms

    in OAI (official action indicated)

    status for a long time,” Cosgrove

    commented at the PDA/FDA con-

    ference. He emphasized the impor-

    tance of complete documentation

    of operations to demonstrate com-

    pliance with GMPs. He also noted

    that documentation by itself “is not

    enough” to demonstrate full com-

    pliance and that FDA inspectors are

    being trained to do a “deeper dive”

    into actual production practices.

    FDA also seeks to halt violative

    imports more quickly by de-linking

    import alerts from warning letters.

    Expeditious action against non-

    compliant imports is important,

    Cosgrove pointed out, because many

    of these products raise data integ-

    rity issues, including data that have

    been deleted, back-dated, copied,

    and fabricated. FDA is highlighting

    data-integrity failures because such

    problems also are linked to GMP vio-

    lations and other problems that rep-

    resent “real risk to patients.”

    OMQ also is looking hard at

    contract manufacturers and how

    well their pharma clients moni-

    tor contract operations for quality

    and compliance. Clients need “to

    get out there,” perhaps put a person

    in the plant, to uncover GMP and

    compliance problems “before we do,”

    Cosgrove advised. He noted that the

    manufacturer holding the approved

    license for a medical product is

    responsible for ensuring quality at all

    its production facilities—including

    those overseas or operated by part-

    ners and suppliers.

    Amidst all these organizational

    changes, FDA is developing a new

    model for assessing plant operations

    based on standardized measures of a

    facility’s state of quality and compli-

    ance. The New Inspection Protocol

    Project (NIPP) will apply to pre-

    approval, GMP surveillance, and for-

    cause inspections. CDER’s Office of

    Pharmaceutical Quality is develop-

    ing the new protocols and planning

    pilot NIPP inspections with ORA.

    The aim is to obtain quantitative

    scores that can help compare sites,

    while also reducing variability in

    observations by different inspectors

    and providing manufacturers with a

    clearer idea of what they need to do

    to maintain quality. While continu-

    ing to document observed deficien-

    cies, inspections also will identify

    practices that exceed basic compli-

    ance requirements to reward positive

    behaviors.

    gloBal CollaBoraTionEfforts at home to develop met-

    rics for evaluating manufacturing

    operations and to streamline and

    target inspections also are being

    applied to foreign manufacturers

    producing medical products for the

    United States. Because FDA lacks the

    resources to monitor the growing

    global pharmaceutical market, US

    and European Union officials are

    looking for greater “mutual reliance”

    on each other’s inspection reports.

    US officials have explored such

    options for more than a decade, only

    to be stymied by legal requirements

    and confusing goals. Now authorities

    are renewing efforts to reduce the

    number of inspections conducted

    by FDA investigators in the EU, and

    by European inspectorates in the US,

    to better target resources to areas of

    greater risk, explained Dara Corrigan,

    FDA associate commissioner for

    global regulatory policy, at the PDA/

    FDA conference.

    FDA conducts thousands of for-

    eign inspections each year, many

    in Europe, Corrigan pointed out,

    and reliable information indicat-

    ing that a facility meets GMPs and

    is a low-risk operation could help

    avoid unnecessary site visits. To

    move forward with a mutual reli-

    ance initiative, FDA investigators

    are observing audits of EU inspec-

    torates, which are conducted by

    other EU member states as part

    of their own internal mutual reli-

    ance inspection program. At the

    same time, EU officials are audit-

    ing ORA district operations to sup-

    port increased EU reliance on FDA

    inspection practices and reports.

    Corrigan noted that FDA offi-

    cials have been impressed with the

    high level of discussion taking place

    during these audits, but a num-

    ber of important issues have to be

    addressed for the initiative to move

    forward. One is that US law requires

    FDA inspection reports to redact

    trade secret information before being

    shared with other regulatory authori-

    ties, a policy that rankles EU officials.

    And while the vast majority of reg-

    istered European drug facilities and

    imported products come from six

    EU member states (Germany, France,

    Italy, United Kingdom, Spain, and

    Ireland), it’s not clear if a mutual

    reliance program could be lim-

    ited to those countries. The path

    forward, Corrigan said, involves

    assessing the variability of EU

    inspectorates and their expertise.

    This is a high priority for both FDA

    and the EU, and, Corrigan stressed,

    “we want to succeed.”

    referenCe 1.J. Wechsler, Pharma. Techn. 39 (5)

    (2015). ◆

    ES699849_BP1115_009.pgs 11.04.2015 00:12 ADV blackyellowmagentacyan

  • 10 BioPharm International www.biopharminternational.com November 2015

    Perspectives on Outsourcing

    Do

    n F

    arr

    all/G

    ett

    y Im

    ag

    es

    Biomanufacturing efficiency is on every-

    one’s minds, being the single most

    important area of focus for global bio-

    processing. And contract manufacturing orga-

    nizations (CMOs) are on the leading edge as

    they implement performance improvements.

    CMOs must remain efficient if they are to be

    competitive—so this is no surprise. Results

    from BioPlan Associates’ 12th Annual Report

    and Survey of Biopharmaceutical Manufacturing

    Capacity and Production (1) offer some clues as

    to what CMOs are doing to remain competitive.

    CMOs’ love affair with single-use devices

    has been well documented. Indeed, single-use

    implementation and integration is a much larger

    focus for CMOs than it is for biotherapeutic

    developers. And as the results in Figure 1 indi-

    cate, it’s easy to see why: nine out of 10 CMOs

    agree that biomanufacturing improvements over

    the past year are coming from the use of dispos-

    able and single-use devices.

    Given that CMOs have long been at the fore-

    front of single-use adoption, it’s perhaps more

    interesting to look at factors that are rising in

    importance for CMOs. One such factor is better

    process development, cited by 81.8% of CMO

    respondents as contributing to improved bioman-

    ufacturing performance, up from

    two-thirds of respondents in 2014.

    This is a notable result, as pro-

    cess development outsourcing has

    been on the rise in recent years.

    Separately, 43% of industry respon-

    dents reported outsourcing at least

    some upstream process develop-

    ment activities to some degree,

    up from just 17.1% back in 2010.

    Additionally, 41% reported at least

    some outsourcing downstream pro-

    cess development activities to some

    degree. Improvements in process

    development, therefore, are an encouraging

    sign for CMOs as this becomes a growing busi-

    ness opportunity.

    A similar pattern plays out in validation ser-

    vices. This is also a growing area of opportunity

    for CMOs, with validation services a more pop-

    ular outsourcing activity than process develop-

    ment. In the 2015 survey, for example, almost

    three-quarters (73%) of industry respondents

    reported outsourcing at least some validation

    services, up from less than two-thirds in 2010.

    Another area to which more CMOs attri-

    bute internal performance improvements is

    upstream production operations. In the 2015

    survey, 64% of respondents said that these

    improvements contributed to better overall

    performance, up from 56% in 2014. In fact,

    CMOs were almost as likely to credit upstream

    improvements as downstream improvements

    with better biomanufacturing performance.

    That may partly be due to the current bottle-

    necks being experienced in purification and

    separation operations. And CMOs’ experience

    with multiple products and campaigns provide

    them expertise that in-house manufacturers

    may not have.

    Upstream biomanufacturing operations out-

    sourcing has been growing more rapidly than

    downstream operations, according to BioPlan’s

    data. In the space of five years, the percentage

    of industry respondents outsourcing upstream

    operations has doubled, from 21% in 2010 to

    42% in 2015. While outsourcing of downstream

    operations has been on the rise, it hasn’t had

    quite the same growth trajectory, up from 28%

    in 2010 to 39% of respondents in 2015.

    Upstream operational improvements are less

    of an industry focus for both CMOs and in-

    house manufacturers. Indeed, when BioPlan

    surveyed the industry on the single most

    important area or operational focus in 2015,

    CMOs Continue to Improve Overall Biomanufacturing Performance Better process development is creating industry benchmarks for bioprocessing.

    Eric Langer is president of BioPlan Associates,

    tel. 301.921.5979, elanger@

    bioplanassociates.com.

    ES699850_BP1115_010.pgs 11.04.2015 00:12 ADV blackyellowmagentacyan

  • For US inquiries, please contact [email protected] • For Asia Pacifi c inquiries, please contact infoAsiaPacifi [email protected]

    For EU and other international inquiries, please contact [email protected]

    Answers that work

    www.vetter-pharma.com

    Looking for a syringe systemfor your sensitive compound?

    Today’s complex compounds developed by pharma and biotech companies require

    new, innovative syringe concepts. Vetter-Ject® offers a syringe closure system

    specially designed for sensitive compounds. By combining a baked-in siliconization

    process with a staked needle, Vetter-Ject® provides a number of signifi cant advantages:

    � Product integrity

    Tamper-evident seal supports product integrity

    � Ready-to-use convenience

    Staked needle offers easy handling and administration

    � High product security

    Baked-in siliconization process reduces product-silicone interaction

    To learn more visit www.vetter-ject.com

    ES700026_BP1115_011_FP.pgs 11.04.2015 02:41 ADV blackyellowmagentacyan

  • 12 BioPharm International www.biopharminternational.com November 2015

    Fig

    ure

    co

    urt

    esy o

    f a

    uth

    or

    Perspectives on Outsourcing

    not a single CMO pointed to

    upstream processing advances as

    the top area; because downstream

    production (DSP) operations issues

    remain strong. A worrisome 64%

    of CMOs said that downstream

    processing is impacting capacity

    and overall production by caus-

    ing at least some bottleneck prob-

    lems (noted by 64%). In fact, only

    a quarter of respondents are cur-

    rently enjoying no bottlenecks in

    their downstream processing.

    CMOs sPending tO Offset POtentiAl CAPACity CrunChNot surprisingly, CMOs are seeing

    more problems than biotherapeutic

    companies due to downstream pro-

    cessing, and are experiencing more

    significant production capacity con-

    straints, too. The BioPlan study indi-

    cated they will likely continue to

    invest in better DSP technologies

    as a way out of these problems, for

    example.

    Facility constraints are the most

    common factor CMOs cite as creat-

    ing capacity crunches at their facil-

    ities over the next five years (cited

    by more than two-thirds). Probably

    by no coincidence, 7 in 10 CMOs

    plan to increase their spending on

    new facility construction this year,

    by an average amount of 11.3%. 

    The next biggest culprit in

    projected capacity constraints is

    downstream purification capacity.

    Spending plans for CMOs are posi-

    tive: almost three-quarters would

    be increasing their capital equip-

    ment budgets, with an average

    increase of 11.7%.

    Expected budget hikes—for cap-

    ital equipment (11.7%) and new

    facility construction (11.3%)—were

    the largest of all areas tracked. To

    grow their businesses, CMOs are

    dedicating funds to offset potential

    capacity constraints in the future.

    Not surprisingly, better down-

    stream purification technologies are

    also on the agenda. CMOs note that

    downstream innovation is the lead-

    ing way to avoid future capacity con-

    straints. Spending projections aren’t

    quite as buoyant for downstream

    innovation, though they are solid.

    In 2015, 6 in 10 will increase spend-

    ing on new technologies to improve

    efficiencies and costs for downstream

    production, for an average budget

    increase of 6.1%. This is likely due

    to new technologies providing more

    incremental increases in efficiencies

    as opposed to new equipment that

    can quickly provide access to more

    capacity and avoid crunches.

    COnClusiOnSingle-use equipment is help-

    ing CMOs achieve performance

    improvements, both for down-

    stream purification and for man-

    ufacturing productivity overall.

    But CMOs are taking numerous

    other factors into account as they

    improve efficiencies and lower costs.

    These range from better analytical

    testing and product release services

    to better operations staff training,

    optimized media and improved

    existing quality management sys-

    tems. Better process development

    is also a growing area of interest for

    CMOs as they take on more process

    development work—both upstream

    and downstream—for clients.

    Nevertheless, one of the main

    routes to overall productivity

    improvements for CMOs will be

    better downstream operations.

    Besides the use of disposable equip-

    ment, a majority of CMOs are

    developing downstream processes

    with fewer process steps. Many are

    also using or evaluating a number

    of technologies, including:

    • Membrane-based filtration tech-

    nologies

    • Ion-exchange membrane tech-

    nologies

    • Ion-exchange technologies with

    higher capacity.

    Biotherapeutic developers might

    keep a close eye on these activi-

    ties. CMOs, with their broad expe-

    rience, multiple product lines, and

    need for rapid changeovers, are

    often at the forefront of innova-

    tion. Though their requirements

    clearly differ from those of bio-

    therapeutic developers, the process

    improvements sparked by innova-

    tions and adopted by CMOs can

    provide a recipe for the industry as

    a whole. As such, it will be interest-

    ing to monitor the activities and

    technologies that CMOs adopt to

    improve downstream production

    operations and overall biomanu-

    facturing productivity.

    referenCe 1. BioPlan Associates, 12th Annual Re-

    port and Survey of Biopharmaceutical Manufacturing Capacity and Produc-tion (Rockville, MD, April 2015), www.bioplanassociates.com/12th, accessed Oct. 12, 2015. ◆

    Figure 1: Improving biomanufacturing performance for CMOs, 2015 v. 2014

    (select responses).

    Use of disposable/single-use devices

    Better process development

    Overall better control of process

    Improved downstream production operations

    Better analytical testing & product release services

    Improved upstream production operations

    86.4%

    83.3%

    81.8%

    66.7%

    68.2%

    77.8%

    68.2%

    72.2%

    63.6%

    66.7%

    63.6%

    55.6%

    2015 2014

    Source: 12th Annual Report and Survey of Biopharmaceutical Manufacturing, April 2015, www.bioplanassociates.com/12th

    ES699851_BP1115_012.pgs 11.04.2015 00:12 ADV blackyellowmagentacyan

  • �bzena adds GMP manufacturing capabiliƟ es.

    Composite CHO™ to produce high

    expressing cell lines for manufacture

    Composite Protein™ technology for

    deimmunisaƟ on of therapeuƟ c proteins

    �omposite ,uman �nƟ bodyΡ technology

    to humanise and deimmunise anƟ bodies

    iTope and TCED™ to idenƟ fy d cell epitopes

    using a proprietary database and algorithm

    EpiScreen™ for ex vivo assessment of the

    immunogenicity of biopharmaceuƟ cals

    Sa

    n D

    ieg

    o,

    USA

    Cambridge, UK

    Coventry, UK

    ThioBridge™ for the generaƟ on of more

    homogeneous anƟ body drug conjugates

    TheraPEG™, CyPEG™ and HiPEG™ for

    linking polymers to therapeuƟ c proteins to

    edžtend their duraƟ on of acƟ on

    PolyPEG™ a low viscosity polymer to

    make it easier to manufacture and inject

    long acƟ ng therapeuƟ c proteins

    GMP manufacturing that specializes in single-use

    technology for the producƟ on of biopharmaceuƟ cals

    for Phase I and Phase II clinical trials

    Process development focuses on improving

    yield at each stage of development by

    opƟ mizing each step in the process

    Biosimilar cell line development in CHO,

    NS0 and SP2/0 including enhanced PQA

    dhe �bzena group has undergone a signiĮ cant transformaƟ on in past few years following

    the acƋuisiƟ on of �nƟ tope in ϮϬϭϯ and PaciĮ cGMP in ϮϬϭϱ. �ringing together these

    companies allows �bzena to support customers projects seamlessly from lead selecƟ on

    through to GMP manufacturing for clinical trials.

    Start your project today. Visit www.abzena.com

    EŽǁ�ĞŶĂďůŝŶŐ�ďĞƩ�Ğƌ�ďŝŽƉŚĂƌŵĂĐĞƵƟ�ĐĂůƐ�ĨƌŽŵ�ŵŽƌĞ�ůŽĐĂƟ�ŽŶƐ�

    R&D site for developing and synthesing

    conjugaƟ on and P�GylaƟ on reagents

    ES700028_BP1115_013_FP.pgs 11.04.2015 02:41 ADV blackyellowmagentacyan

  • 14 BioPharm International www.biopharminternational.com November 2015

    PL

    AIN

    VIE

    W/M

    aria T

    outo

    ud

    aki/G

    ett

    y Im

    ag

    es;

    Dan W

    ard

    Whe t he r o ut s o u r c i n g

    aseptic techniques to a

    third party, or perform-

    ing these tasks in an

    academic setting or in an in-house

    laboratory, certain tools, technolo-

    gies, and standard operating proce-

    dures are necessary to ensure sterility

    across settings. Because many biolog-

    ics cannot be terminally sterilized,

    isolators and restricted access barrier

    systems (RABS) are typically the go-to

    tools manufacturers use to ensure

    product sterility.

    To gain some insight into how to

    best prepare sterile, parenteral prod-

    ucts, BioPharm International spoke to

    experts in both the theory and the

    practice of sterile drug preparation.

    Specifically, the publication spoke to

    Bivash Mandal, PhD, a senior research

    specialist at the Plough Center for Sterile

    Drug Delivery Systems in the University

    of Tennessee Health Science Center,

    and Bernd Stauss, senior vice-president

    of production/engineering at Vetter

    Pharma-Fertigung GmbH & Co.

    The Plough Center for Sterile Drug

    Delivery Systems announced in August

    2015 that it is installing three PODs

    from G-CON Manufacturing in a new

    facility on campus to manufacture

    drugs for sponsors and train profession-

    als on cGMPs for the large-scale produc-

    tion of pharmaceuticals (1). Although

    the location currently has the capacity

    to manufacture small-volume parenteral

    preparations for clinical investigation,

    the facility expansion, which began

    in September 2015, will al low the

    Best practices for sterility assurance in Fill/Finish Operations

    Randi Hernandez

    Two experts discuss best

    practices to achieve

    acceptable sterility

    assurance levels for

    aseptically filled products.

    Fill/Finish

    ES701255_BP1115_014.pgs 11.05.2015 21:58 ADV blackyellowmagentacyan

  • November 2015 www.biopharminternational.com BioPharm International 15

    university to manufacture drugs

    for preclinical and clinical trials.

    The PODs are slated to be up and

    running by 2016.

    Vetter is an outsourcing com-

    pany that has helped guide dozens

    of product approvals for biophar-

    maceutical compounds and spe-

    cializes in the commercial filling

    and packaging of parenteral drugs.

    In the past few years, Vetter has

    focused on innovation in the

    field, combining the advantages

    of isolators and RABS to create a

    new approach in sterility assur-

    ance, which the company calls

    its “Improved RABS Concept.”

    The technique features an accel-

    erated process cycle and an auto-

    mated decontamination function

    for increased operational excel-

    lence in aseptic processes (2).

    EquiPmEnt trEndSBioPharm: What are the trends in

    the use of RABS and isolators? Is

    use of this type of equipment the

    best way to ensure the sterility of

    one’s fill/finish processes?

    Mandal: Aseptic processing is a

    complex manufacturing tech-

    nology that can be achieved by

    using aseptic cleanrooms (manned

    human-scale cleanrooms), isola-

    tors/restricted access barrier sys-

    tems (RABS), or both. As far

    as the industrial trends are con-

    cerned, some firms have taken a

    mix-and-match approach. RABS

    and isolators can be used in the

    manufacture of biologics, includ-

    ing vaccines, gene therapies, and

    protein-based drugs. Often, bio-

    logic products are preservative-free,

    contain growth media, and are

    easily susceptible to contamina-

    tion. Another area that demands

    the use of RABS and isolators is

    the manufacture of sterile drug

    products with toxic, cytotoxic, and

    highly potent molecules, which

    require stringent barriers to pro-

    tect personnel who are handling

    these materials. In general, RABS

    and isolators are being used for

    smaller-volume and high-value

    pharmaceuticals. The benefit/cost

    balance has to be considered when

    discussing the use of barriers: RABS

    and isolators come with a high

    price tag and are associated with

    additional expenses related to the

    operation of a cleanroom, such as

    energy costs, operating costs, test-

    ing costs, and gown costs.

    Because it has been established

    that the personnel working in

    cleanrooms can be a major source

    of contamination, RABS and isola-

    tors are preferred as a means of a

    physical barrier to separate people

    from filling processes. According

    to FDA guidance on aseptic pro-

    cessing, isolators and closed RABS

    are superior in their ability to con-

    trol contamination and reduce

    validation workload. Operators

    must use these advanced tech-

    nologies with caution because the

    use of RABS and isolators alone

    does not guarantee the sterility

    of products. In both isolators and

    RABS, for instance, operators use

    glove ports, and glove ports need

    to be inspected on a daily basis.

    Moreover, gloves are considered

    a primary route of contamina-

    tion, and they are a common cause

    of failure in isolator technology.

    Complete automation and use of

    robotic technology in conjunction

    with isolators and RABS should be

    Fill/Finish

    BEST PRACTICES IN FREEZE/THAW OPERATIONS

    BioPharm International asked Bivash Mandal, PhD, senior research specialist

    at The Plough Center for Sterile Drug Delivery Systems in The University of

    Tennessee Health Science Center, for a few tips to help ensure optimal freeze/

    thaw operations.

    BioPharm: What are the dangers associated with multiple freeze/

    thaw operations?

    Mandal: Multiple freezing and thawing of a biopharmaceutical product

    could affect the chemical and physical properties of the product. In the case of

    protein drugs, the procedure can stress and may irreversibly denature complex

    macromolecular structures, altering their stability. The rate at which freeze/

    thaw processes occur plays a significant role in product quality. Fast freezing

    rates could lead to smaller ice-crystal formation. This process can result in their

    partial unfolding, increased aggregation, and decreased biological activity. There

    is also an increased risk of the entrapment of air during fast freezing, which can

    denature proteins as air-liquid interfaces form. On the other hand, slow thawing

    rates often result in ice recrystallization, and the shear stress generated by slow

    freezing can damage biologics.

    BioPharm: How does the geometry of vials or cryobags affect the fill/finish

    process of allogeneic cells?

    Mandal: For the fill/finish of allogeneic cells, one of the crucial steps is the

    final freezing step for cryopreservation of the cells with an acceptable shelf life.

    An optimal cooling rate is one of the critical parameters affecting the survival of

    cells during cryopreservation. For cryovials, freezing patterns will be influenced

    by the variation in container-base geometry. If a vial’s base is not flat and does

    not have a uniform thickness, there may be uneven thermal contact between

    a sample and the lyophilization shelf. The mechanism of heat exchange will be

    affected based on the dimensions and geometry of the sample container and

    whether the container rests directly on a shelf or is supported in a tray.

    —Randi Hernandez

    ES700988_BP1115_015.pgs 11.05.2015 17:53 ADV blackyellowmagentacyan

  • 16 BioPharm International www.biopharminternational.com November 2015

    developed to eliminate the human

    interventions that are performed

    using glove/sleeve assemblies.

    Stauss: There are two distinct

    technologies dominating the fill/

    finish process: isolators and RABS.

    Each technology has its advantages.

    With isolator technology, the pro-

    cessing takes place in systems that

    are entirely shut off from the outside

    environment. As it pertains to steril-

    ity assurance levels (SAL), isolators

    are often considered the best solu-

    tion due to the automatic decontam-

    ination processes involved. However,

    isolators need extensive decontam-

    ination and preparation processes

    following a batch to enable a safe

    change in product.

    RABS technology also achieves the

    SAL currently required by regulatory

    authorities. With this technology,

    the physical barriers of a production

    plant are limited; a RABS requires

    installation in a higher-class envi-

    ronment (at least ISO 7, with the

    RABS located in an ISO 5 area).

    Conversely, this system provides

    flexibility and high-capacity utiliza-

    tion for multi-product filling lines;

    this is a reason why RABS are often

    found at CDMOs [contract develop-

    ment and manufacturing organi-

    zations]. When choosing between

    isolator and RABS technology, each

    company has to make the decision

    that best fits their production situa-

    tion and needs.

    BioPharm: What equipment is

    common for those performing fill/

    finish operations?

    Mandal: For fill/finish operations,

    liquid-filling equipment (manual/

    semiautomatic/automatic), peristal-

    tic pumps, filtration apparatuses,

    a lyophilizer (if required), a vial/

    ampoule sealer/crimper (semiauto-

    matic/automatic), and a biosafety

    cabinet (hood) are required. During

    fill/finish operations, it is also

    required to monitor the environ-

    mental air quality by passive sam-

    pling using settling plates and active

    sampling using a centrifugal sam-

    pler and an impactor-type sampler.

    A laser particle counter can moni-

    tor the total particulate count of the

    environmental air.

    Successful product

    integrity testing

    using deterministic

    or probabilistic

    methods is the basis

    for enabling sterility

    in manufactured drug

    products.—Bernd

    Stauss, Vetter

    quAlity mEASurEmEntSBioPharm: What have been some

    common performance gaps when it

    comes to environmental monitoring?

    Mandal: Some of the common

    performance gaps in environmen-

    tal monitoring include not follow-

    ing standard operating procedures,

    not monitoring in all aseptic pro-

    cessing areas, inadequate corrective

    actions, not responding in a timely

    fashion to out-of-limit results,

    inadequate personnel training,

    failure to validate the cleaning and

    sanitization procedures, failure to

    trend environmental monitoring

    data, failure to identify common

    microorganisms, and inadequate

    documentation of deviations.

    BioPharm: How are aseptically

    manufactured drug products best

    evaluated for their sterility?

    Stauss: Proving the sterility of

    manufactured drug products is

    crucial to a drug manufacturer.

    In the first step, the design of the

    applied primary packaging materi-

    als needs to meet integrity require-

    ments. Successful product integrity

    testing using deterministic or prob-

    abilistic methods is the basis for

    enabling sterility in manufactured

    drug products. After the integrity

    of the package design is estab-

    lished, incoming packaging mate-

    rials are routinely tested to ensure

    they meet specifications.

    Equipment surfaces that come

    into contact with sterilized drug

    product or sterilized primary pack-

    aging materials, as well as any cru-

    cial equipment in the cleanroom,

    needs to be sterilized by using vali-

    dated sterilization methods. Moist-

    heat and dry-heat sterilization are

    the most commonly used steril-

    ization methods. Furthermore, the

    aseptic processing operations need

    to be tested for their ability to pro-

    duce sterile products via process sim-

    ulations (media fill). During media

    fill, microbiological growth medium

    is exposed to product contact sur-

    faces to simulate the exposure that

    the product may undergo during

    manufacturing. The sealed contain-

    ers filled with the medium are then

    incubated at defined temperatures to

    detect microbial contamination.

    During manufacturing, varying

    controls like bioburden and endo-

    burden testing of product and fil-

    ter integrity testing are performed.

    Another important aspect is the

    environmental monitoring of the

    surroundings. Before release of a

    batch, a sterility test in an isola-

    tor is performed to further demon-

    strate sterility of the filled batch.

    Mandal: Aseptically manufac-

    tured drugs must be sterile, pyro-

    gen-free, particulate-free, stable,

    and isotonic. Sterility testing must

    be conducted on every batch of

    a product that is manufactured.

    FDA consistently emphasizes that

    sterility testing is to remain a cur-

    rent good manufacturing practice.

    Chapter of the United States

    Fill/Finish

    ES700719_BP1115_016.pgs 11.04.2015 23:38 ADV blackyellowmagentacyan

  • www.gelifesciences.com/BioProcess

    Xcellerex XDR systems offer the benefi ts of single-use technology

    in a stirred-tank bioreactor design. Developed for scalability

    and robustness, the XDR platform provides the performance

    and fl exibility needed from process development to large-scale

    biopharmaceutical manufacturing.

    When you need a fl exible and scalable cell culture solution, we

    are a partner like no other.

    Predictable process scaling.Xcellerex™ bioreactor platform.

    upstream downstream single-use services

    GE, GE monogram, and Xcellerex are trademarks of General Electric Company. © 2014–2015 General Electric Company. First published Oct. 2014.GE Healthcare Bio-Sciences AB. Björkgatan 30, 751 84 Uppsala, Sweden

    29133379 AB 08/2015

    ES700027_BP1115_017_FP.pgs 11.04.2015 02:41 ADV blackyellowmagentacyan

  • 18 BioPharm International www.biopharminternational.com November 2015

    Fill/Finish

    Pharmacopeia (USP) states that ste-

    rility tests on parenteral dosage

    forms are not intended to be used

    as a single criterion for the accept-

    ability of a product (3). Sterility

    assurance is achieved primarily by

    the validation of the sterilization

    processes and the aseptic process-

    ing procedures.

    Aseptically

    manufactured

    drugs must be

    sterile, pyrogen-

    free, particulate-

    free, stable, and

    isotonic.—Bivash

    Mandal, University

    of Tennessee Health

    Science Center

    Ideal ly, every v ia l/syr inge/

    ampoule manufactured must be

    tested for its sterility. Because

    sterility testing is a destructive

    process, however, testing each

    individual unit is not possible. USP

    provides guidance for the

    minimum number of articles that

    need to be tested from each manu-

    factured batch.

    The sterility test can be per-

    formed by two different methods:

    by the direct inoculation method

    or by the membrane filtration

    method. In the direct inoculation

    method, a predetermined amount

    of product is added directly to the

    medium under aseptic conditions

    and incubated. In the membrane

    filtration method, the contents of

    the product to be tested are filtered

    through an appropriate-sized filter,

    such that if any microorganisms

    were to be present, they would be

    retained on the filter. This filter is

    then washed with specified solutions

    to remove any retained product, and

    finally, the filter is incubated with

    medium at appropriate conditions

    for at least 14 days.

    Two different media must be

    used for testing, irrespective of the

    testing method used. Fluid thiogly-

    collate medium (FTM) is used to

    culture primarily anaerobic micro-

    organisms, although it can support

    the growth of aerobic microorgan-

    isms as well. Trypticase soy broth

    (TSB), also called the soybean

    casein digest medium, is used to

    test for the presence of fungi and

    aerobic microorganisms. If a par-

    ticular drug product inhibits the

    growth of bacteria, such as is the

    case with beta-lactam antibiotics,

    the formulation of the medium

    can be modified to include cer-

    tain agents that can deactivate the

    antibiotics, such as beta-lactamase.

    Alternatively, the membrane filtra-

    tion method can be used.

    A failure of the sterility test is

    indicated by a growth in one or

    more of the incubated samples.

    There is no such thing as a false

    positive in the sterility testing of

    an aseptically manufactured prod-

    uct. A comprehensive written inves-

    tigation follows, which includes

    identification of the bacteria,

    specific conclusions, and correc-

    tive actions. A sterility test that is

    positive may be indicative of pro-

    duction, personnel, or laboratory

    problems. The most commonly

    found microorganisms in steril-

    ity test failures include, but are not

    limited to: Staphylococcus aureus,

    Pseudomonas aeruginosa, Escherichia

    coli, Enterobacter aerogenes, Neisseria

    gonorrhoeae, Aspergillus niger, and

    Candida albicans.

    Fill/FiniSh BESt PrActicESBioPharm: Can you describe some

    best practices for decontamination?

    Stauss: The goal of a service pro-

    vider to the biopharmaceutical

    industry is to provide its custom-

    ers with reliable and efficient asep-

    tic production processes, which

    are supported by safe and effec-

    tive cleaning and decontamination

    processes.

    Automated decontamination of

    RABS reduces downtime, increases

    capacity utilization, and improves

    overall equipment effectiveness.

    Prior to the start of the decontami-

    nation process, format parts are

    cleaned offline, in full, and auto-

    matically to remove particles, sili-

    con, or residues, for example. This

    automated cleaning process rep-

    resents an important advantage

    as compared to isolators, where a

    manual cleaning process is nor-

    mally applied.

    Mandal: As an alternative to

    formaldehyde-based sterilization,

    vaporized hydrogen peroxide (VHP)

    was introduced in the mid-1980s

    to clean and decontaminate equip-

    ment and machinery in the health-

    care industry. Since then, the use of

    VHP has been steadily increasing

    due to the following advantages:

    •Efficacyinrapid

    decontamination of machines at

    ambient temperatures and low

    concentrations

    •Stronghistoryofuseand

    positive efficacy data on a broad

    range of bacteria, fungus, spores,

    and viruses

    •Provenefficacytestingwith

    biological indicators and

    chemical indictors

    •Abilitytokillresistantspores

    •Usewithinacontrolledprocess

    with real-time concentration

    monitoring

    •Notoxicbyproductswithits

    use (VHP is a green solution)

    •Associatedwithlessexposurerisk

    to personnel and products outside

    of a decontamination zone

    ES700727_BP1115_018.pgs 11.04.2015 23:38 ADV blackyellowmagentacyan

  • November 2015 www.biopharminternational.com BioPharm International 19

    •Afavorablesafetyprofile

    (Typical concentrations used

    are 150–700 ppm as compared

    with formaldehyde [8000–10000

    ppm] and chlorine dioxide

    350–1500 ppm)

    •Nolengthyaerationperiod

    •Noresidue

    •Astrongmaterialand

    component compatibility profile

    •Registeredbythe

    Environmental Protection

    Agency (EPA)

    •ApprovedbyFDA.

    casE studiEsBioPharm: Can you describe some

    of your most challenging fill/finish

    projects and what you did to over-

    come obstacles that were presented?

    Mandal: The Plough facility at

    the University of Tennessee has

    been manufacturing small-scale

    batches for preclinical and Phase

    I clinical trials for sponsors. We

    have been using an aseptic clean-

    room with manual intervention

    and semiautomatic filling lines.

    Most of the challenges we have

    faced were mechanical or instru-

    ment-oriented.

    One of the projects (manufacture

    of a sterile solution of polysaccha-

    ride) had issues with the filling line

    clogging when the filling opera-

    tion was halted to switch person-

    nel. The formulated product was

    good, however, and was still within

    acceptable limits of viscosity. Upon

    investigation, we found that resid-

    ual solution—which is in contact

    with the filling needle tips—evapo-

    rated in the laminar flow. We were

    unable to remove the clot with high

    pressure. The problem was solved

    by running the entire fill continu-

    ously, without interruptions.

    Another challenge was with a

    project focused on a parenteral that

    was made up of an oily solution.

    The process required us to overlay

    nitrogen to protect the product

    from oxidation. After stoppering

    the product, the vial stopper even-

    tually became pushed out in time.

    The solution to the problem was

    to crimp the vial in a reasonable

    amount of time after stoppering.

    Recently, we had a project on the

    preparation and aseptic fill/finish

    of a liposomal product contain-

    ing a cytotoxic chemotherapeutic.

    Liposomal products are notoriously

    challenging fill/finish projects

    because of issues with filtration,

    drug loading, filter compatibil-

    ity, and particle-size distribution.

    Compatibility of the filter was an

    important issue due to the drug

    being adsorbed in the filter. The

    proper control of the filtration

    pressure was crucial, because there

    is an increased occurrence of drug

    loss from liposomes during filtra-

    tion at higher pressures.

    Additionally, the containment

    of the cytotoxic chemotherapeu-

    tic proved challenging. Special

    procedures should be adopted to

    deactivate the drug contaminated

    materials after fill/finish. Cleaning

    validation of the equipment should

    be conducted in order to obviate

    cross-contamination.

    Stauss: Based on our day-to-day

    experiences in customer projects, we

    see the overall market is increasingly

    becoming more challenging, par-

    ticularly in areas such as:

    • An increase in high-value

    products in smaller batch sizes

    • The cont inuous increase

    in regulatory requirements,

    including anticounterfeiting

    activities

    • Ever-more complex supply

    chains on the customer side,

    which have resulted in more

    compl ic ate d r e que s t s fo r

    CDMOs.

    High-value products are often

    based on complex compounds.

    They demand high accuracy on the

    filling line and have an increased

    sensitivity to manufacturing pro-

    cesses and environmental condi-

    tions. A good example of a difficult

    fill/finish project is the handling of

    a highly sensitive API that requires

    very small fill volume in a syringe.

    Small filling volumes in such cir-

    cumstances create signif icant

    demands on all production areas,

    including process design, technical

    equipment, and packaging mate-

    rial. This, in turn, creates high

    demands on the operating staff.

    In such cases, packaging material

    and processes need to be adapted to

    meet the requirements of a product.

    Using the correct application tech-

    nique of the silicone coating on a

    syringe is a good example of a com-

    mon packaging challenge.

    Comprehensive project manage-

    ment is necessary to handle such

    a project successfully, taking into

    consideration the needs of both the

    product and the customer. To pro-

    actively enable a successful product

    launch, every potential impediment

    to the best outcome in fulfilling

    product requirements—including

    manufacturing processes, use of tech-

    nical equipment, and proper staffing,

    to name a few—must be taken into

    account during the project phase.

    rEFErEncEs 1. The University of Tennessee Health

    Science Center, “New Plough

    Center for Sterile Drug Delivery

    Systems to Expand UTHSC’s

    National and Global Position as

    a Pharmaceutical Manufacturer,”

    Press Release, http://news.uthsc.

    edu/new-plough-center-sterile-

    drug-delivery-systems-expand-

    uthscs-national-global-position-

    pharmaceutical-manufacturer/,

    accessed Oct. 13, 2015.

    2. Vetter, “Vetter Embarks on a 300

    Million Euro Investment Strategy

    for Further Development to its

    Manufacturing Sites and to Make

    Available Additional Manufacturing

    Capacities,” Press Release, www.

    vetter-pharma.com/en/newsroom/

    press/publications/vetter-embarks-

    on-a-300-million-euro-investment-

    strategy-for-further-development-to-

    its-manufacturing-sites-and-to-make-

    available-additional-manufacturing-

    capacities/vetter-embarks-on-a-300-

    million-euro-investment-strategy/,

    accessed Oct. 13, 2015.

    3. USP, USP General Chapter ,

    “Sterility Tests,” USP 29–NF 24

    (US Pharmacopeial Convention,

    Rockville, MD, 2006). ♦

    Fill/Finish

    ES701280_BP1115_019.pgs 11.05.2015 22:41 ADV blackyellowmagentacyan

  • 20 BioPharm International www.biopharminternational.com November 2015

    Med

    icalR

    F.co

    m/G

    ett

    y Im

    ag

    es

    This article reviews the impli-

    cations of cell-culture con-

    ditions on biologic product

    quality, focusing on glycosyl-

    ation and analytical techniques for its

    accurate assessment. Glycosylation can

    potentially affect a protein’s half-life,

    immunogenicity, binding activity, and

    stability. It is a complex process that

    consists of the attachment of carbohy-

    drate moieties, with possible attach-

    ment sites via asparagine (N-linkage)

    or serine/threonine (O-linkage) amino

    acids in protein structures. In mam-

    malian cell culture processes, the use

    of different species can potentially

    produce significant differences in the

    types of glycosylation that can occur.

    These differences in glycosylation can

    have significant effects on the quality

    of the therapeutic protein produced, as

    can the choice of cell clone, the basal

    and feed media used, and the cell-

    culture conditions.

    The choice of host cell and the bioreac-

    tor conditions used in bioproduction of

    proteins significantly affects protein prod-

    uct quality. This is due both to the struc-

    tural complexity of proteins themselves

    and also to species-specific post-transla-

    tional modifications that may occur dur-

    ing the cell-culture process, glycosylation

    being of particular importance.

    Protein theraPeutics and cell-culture effects on Protein qualityBiopharmaceutical drugs are proteins

    with polymeric structures, built up

    in a series of structural levels starting

    implications of cell culture conditions on Protein Glycosylation

    Richard Easton and Michiel E. Ultee

    The authors present a

    review of the techniques commonly

    used for glycosylation

    analysis.

    Michiel E. Ultee, PhD, is principal

    at ulteemit Bioconsulting, llc, and

    Richard Easton, PhD, is team

    leader, carbohydrate analysis,

    sGs life science services.

    upstream Processing

    ES700355_BP1115_020.pgs 11.04.2015 18:14 ADV blackyellowmagentacyan

  • EMD Millipore Corp. is a subsidiary of Merck KGaA, Darmstadt, Germany

    Your fast track through regulatory challenges.The new Emprove® program. Does the constantly changing regulatory landscape sometimes feel like a maze? The new Emprove® program provides the answers you need, with a portfolio of 400 pharma raw and starting materials backed by information to support your qualification, risk assessment, and process optimization activities.

    • Portfolio of products to address different risk levels• Elemental Impurity Information (ICH Q3D) • Online access to all dossiers in the new Emprove® Suite

    Take advantage of this process accelerating combination of high-quality products and targeted insight. We help you find the fast track through the maze.

    Find out how at:www.emdmillipore.com/emprove

    EMD Millipore, the M mark and Emprove are registered trademarks of Merck KGaA, Darmstadt, Germany.

    © 2015 EMD Millipore Corporation, Billerica, MA, SA. All rights reserved.

    ES700057_BP1115_A21_FP.pgs 11.04.2015 02:42 ADV blackyellowmagentacyan

  • Merck Millipore is a business of

    Your fast track through regulatory challenges.The new Emprove® program. Does the constantly changing regulatory landscape sometimes feel like a maze? The new Emprove® program provides the answers you need, with a portfolio of 400 pharma raw and starting materials backed by information to support your qualification, risk assessment, and process optimization activities.

    • Portfolio of products to address different risk levels• Elemental Impurity Information (ICH Q3D) • Online access to all dossiers in the new Emprove® Suite

    Take advantage of this process accelerating combination of high-quality products and targeted insight. We help you find the fast track through the maze.

    Find out how at:www.merckmillipore.com/emprove

    Merck Millipore, the M mark and Emprove are registered trademarks of Merck KGaA, Darmstadt, Germany.

    © 2015 Merck KGaA, Darmstadt, Germany. All rights reserved.

    ES700042_BP1115_B21_FP.pgs 11.04.2015 02:42 ADV blackyellowmagentacyan

  • 22 BioPharm International www.biopharminternational.com November 2015

    AL

    L F

    IGU

    RE

    S A

    RE

    CO

    UR

    TE

    SY

    OF

    TH

    E A

    UT

    HO

    RS

    from the amino-acid sequence,

    referred to as primary structure,

    through folding of the amino

    acid chains into local (second-

    ary) and longer-range (tertiary)

    three-dimensional conforma-

    tions. Multi-chain proteins, such

    as IgG antibodies, additionally

    have a quaternar y st r uc ture

    resulting from structural associa-

    tions between the subunits.

    the choice of host cell

    and the bioreactor

    conditions used in

    bioproduction of

    proteins significantly

    affects protein

    product quality.

    The choice of host-cell line for

    recombinant protein production

    depends first on the protein’s

    molecular properties. Certain bac-

    teria can be used for production of

    the simplest proteins, those that

    are composed only of amino-acid

    polymers, with no post-transla-

    tional modifications (PTMs) such

    as glycosylation, because most

    bacterial strains are incapable of

    glycosylation. Production is fast

    using simple media; however, puri-

    fication can be challenging. Rapid

    production of proteins with primi-

    tive glycosylation can be achieved

    using yeast. Insect cells, generally

    used with a baculovirus vector in

    transient fashion, are used mostly

    for R&D and niche products.

    Mammalian cells are used for the

    production of complex proteins

    such as antibodies and enzymes,

    requiring full PTMs, including the

    production of complex carbohydrates.

    Proteins are delicate molecules

    compared with small-molecule

    drugs and present multiple stabil-

    ity challenges. A typical glycopro-

    tein such as an IgG antibody has

    many sites of variability within its

    structure, which comprises four

    chains with a total molecular

    weight of 150,000 Da. Additionally,

    there are several post-translational

    modifications of the protein chain

    that can occur, such as oxidation

    and deamidation of specific amino

    acids. Each heavy chain also

    includes a site where glycosylation

    takes place (Figure 1).

    Why is Glycosylation imPortant?Many complex proteins such as

    antibodies and enzymes are glyco-

    proteins, containing from 2–30%

    carbohydrate. Glycosylation is a

    complex process, with the carbo-

    hydrate attached to the protein

    either via the amino acids aspara-

    gine (N-linked) or serine/threonine

    (O-linked). Multiple sugar types

    are possible, each with multiple

    attachment sites, and variation of

    the mammalian cells used in pro-

    duction can lead to subtle glycosyl-

    ation differences (1).

    The choice of cell clone affects

    product quality. Each clone has

    slightly different abilities for gly-

    cosylation and other PTMs, and

    viabilities vary, resulting in differ-

    ences in released intracellular deg-

    radative enzymes into the culture.

    Therefore, it may be necessary to

    select a cell clone that is not the

    highest producer to achieve the

    desired protein quality.

    The extent of glycosylation can

    also vary depending on the basal

    and feed media, even with a single

    clone producing a single monoclo-

    nal antibody, or single basal media

    with varied feeds.

    effects of cell-culture conditionsCel l- cu lture parameters that

    affect the type and extent of gly-

    cosylation include pH and CO2levels; the amount of dissolved

    oxygen (dO2); the temperature;

    the levels and types of nutrients;

    the presence and types of gly-

    can precursors; cell viability, as

    dying cells release degradative

    enzymes; and the level of process

    control.

    upstream Processing

    Figure 1: Graphic showing IgG antibody structure, sites for glycosylation, and

    potential variability.

    ES700969_BP1115_022.pgs 11.05.2015 17:41 ADV blackyellowmagentacyan

  • Bioreactors offer much greater

    control of pH and dissolved gasses

    than shake flasks do, and hence,

    better process control, but shake

    flasks are more economical and

    readily allow larger numbers or

    arrays. Shake-flasks are, therefore,

    commonly employed for early

    scouting studies on media and

    feed conditions. Optimum cell-

    culture development is achieved

    by working with bioreactors to

    enhance growth and productiv-

    ity via selection of basal media

    and culture feeds and the timing

    for these feeds; optimizing bio-

    reactor oxygenation conditions,

    such as CO2 levels, pH, agitation,

    temperature, seeding densities,

    and split ratios; extending the

    production phase of cell culture

    through the use of temperature

    shift; and minimizing accumula-

    tion of growth-inhibiting metabo-

    lites such as ammonia.

    More recently, the availabil-

    ity of miniaturized bioreactors

    has provided an effective and

    e f f ic ient way of conduc t ing

    multi-parameter studies. Thus,

    ‘ big- data’ approaches a l low-

    ing design-of-experiment (DoE)

    studies, in which multiple inter-

    reacting bioreactor conditions

    are evaluated simultaneously, are

    made possible. Such studies require

    the employment of automated,

    large-number bioreactor arrays to

    make the process feasible (2).

    sPecial cell-culture considerations for BiosimilarsBiosimilars are “generic” ver-

    sions of protein pharmaceuticals

    that must be highly similar to

    the innovator drug in order to

    be classified as such. Regulatory

    guidelines require extensive ana-

    lytical testing side by side with

    the innovator drug, including

    full glycosylation profiles (3).

    Similarity to the innovator drug

    is paramount; this must begin

    from clone selection and pro-

    ceed throughout process devel-

    opment. Therefore, rather then

    only selecting for clones with

    the highest titer, as is usually the

    case for innovator drugs, selec-

    t ion is f irst for biosimilar ity,

    which may mean that some of

    the highest producing clones are

    not selected.

    Figure 2: Stacked high pH anion-exchange chromatography procedure with

    pulsed amperometric detection (HPAEC–PAD) chromatograms of glycans

    released from three preparations of bovine fetuin.

    125

    100

    75

    50

    25

    -10

    0

    NanoCoulombs(nC)

    1

    2

    3

    13 25 38 50 63 75 88 100 120

    min

    BioPharm International magazine

    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.

    EACH ISSUE INCLUDES:

    r� Quality/Analytics

    r� 6QTUSFBN�1SPDFTTJOH

    r� %PXOTUSFBN�1SPDFTTJOH

    r� 1FFS�3FWJFXFE�5FDIOJDBM�/PUFT

    r� 1SPEVDU�4QPUMJHIU

    r� 1FSTQFDUJWFT�PO�0VUTPVSDJOH

    VISIT OUR WEBSITE TO SUBSCRIBE FOR FREE TODAY! XXX�#JP1IBSN*OUFSOBUJPOBM�DPN

    JOIN OVER 31,000 SUBSCRIBERS

    SIGN UP FOR YOUR FREE SUBSCRIPTION TODAY!BIOPHARM OFFERS PRINT &

    DIGITAL SUBSCRIPTIONS:

    www.BioPharmInternational.com/subscribe

    www.twitter.com/BioPharmIntl

    www.linkedin.com/groups/BioPharm-International-4027200

    ES700968_BP1115_023.pgs 11.05.2015 17:41 ADV blackyellowmagentacyan

  • 24 BioPharm International www.biopharminternational.com November 2015

    upstream Processing

    techniques for Glycosylation analysisN-glycosylation occurs when the

    carbohydrate is attached to aspar-

    agine in the consensus sequence

    asparagine-X-serine/threonine,

    where X is any amino acid except

    proline. As a nascent protein is

    being synthesized in the endo-

    plasmic ret iculum within the

    cell, an en bloc transfer of a pre-

    formed, lipid-anchored conserved

    glycan occurs. As the synthesized

    protein makes its way through

    the Golgi apparatus, the con-

    served N-linked glyan is ‘pro-

    cessed’ by enzymes (glycosidases

    and glycosyltransferases). It is

    the presence of these processing

    enzymes, their relative levels, and

    the accessibility of the glycosyl-

    ation site on the protein to these

    enzymes that determine the final

    glycosylation of a protein.

    O-glycosylation occurs on the

    amino acids serine and threo-

    nine, but not in accordance with

    a l inear consensus sequence.

    There are some rules governing

    the process—for example, there are

    often nearby proline amino acids

    within the regions of glycosyl-

    ation, and also quite often, tandem

    repeats of serine and theonine.

    analysis of n-GlycansAnalysis of N-glycans is the most

    active area of research. The tech-

    niques that apply to analysis

    of N-glycosylation also apply to

    O-glycosylation.

    The most abundant mam-

    malian N-glycan structure is the

    complex type where a number of

    N-acetylglucosamine structures

    are appended to the molecule and

    extended with galactose, fucose, and

    sialic acid residues to give between

    two and four antennal structures.

    The exact structure reflects the

    nature of the enzymes present in the

    cell type used in the expression pro-

    cess as well as the precise environ-

    ment in which the cells are located.

    International Conference on

    Harmonisat ion ( ICH) Qualit y

    Guidelines Q6B describes the type

    of analysis that should be per-

    formed in relation to these struc-

    tures to understand the quantities

    of the different types of monosac-

    charides present, the nature of the

    glycans in terms of their antennary

    profile, and linkage of monosac-

    charides in each structure but also

    the structure of the glycans at the

    different glycosylation sites on the

    protein backbone (4, 5).

    analytical Procedure: the detailsThe analytical procedure of gly-

    cosylation involves a number of

    physical and enzymatic steps to

    release the glycans and then sepa-

    rate them from the peptides and

    any O-glycopeptides present in

    the mixture. O-glycans can be

    released chemically from these

    O-glycopeptides and purified sep-

    arately from the remaining pep-

    tide chains.

    A permethylation derivatization

    procedure is then performed on the

    separated N-glycans and O-glycans,

    enabling them to be analyzed

    using matrix-assisted laser desorp-

    tion ionization–mass spectrometry

    (MALDI–MS) analysis.

    It is also possible to analyze the

    samples chromatographically with-

    out having to employ the permeth-

    ylation procedure. For example, a

    high pH anion-exchange chroma-

    tography procedure with pulsed

    amperometric detection (HPAEC–

    PAD) can be used for the analy-

    sis of N-glycans. An example of

    this can be seen in the N-glycans

    released from bovine fetuin, as

    shown in Figure 2. This method

    gave consistent data across three

    preparations, a series of clustered

    peaks representing di-, tri-, and

    tetra-sialylated glycans (i.e., two,

    three, and four sialic acid groups

    on the N-glycans within each clus-

    ter). The analysis gives some struc-

    tural information in terms of an

    idea of what has been produced

    but little information in terms of

    the precise nature of what the gly-

    cans are. Techniques like this are,

    therefore, useful for comparative

    work but do not give much struc-

    tural information for characteriza-

    tion of the molecules.

    one final step in

    characterization is

    to determine the

    stereospecificity

    of any linkages

    within structures

    wh