preliminary program wcbp 2016...chi-ting huang, cth analytical consulting peter johnson, 3m drug...

156
PRELIMINARY PROGRAM WCBP 2016 20 th Symposium on the Interface of Regulatory and Analytical Sciences for Biotechnology Health Products Symposium Co-Chairs: Michael Kennedy, CBER Juhong Liu, CDER Brian K. Nunnally, Biogen The Mayflower Hotel Washington, DC January 26-28, 2016

Upload: others

Post on 07-Feb-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • PRELIMINARY PROGRAM

    WCBP 2016

    20th Symposium on the Interface of

    Regulatory and Analytical Sciences for

    Biotechnology Health Products

    Symposium Co-Chairs:

    Michael Kennedy, CBER

    Juhong Liu, CDER

    Brian K. Nunnally, Biogen

    The Mayflower Hotel

    Washington, DC

    January 26-28, 2016

  • Table of Contents

  • The Organizing Committee gratefully acknowledges the Symposium Program

    Partners for their generous support of WCBP 2016

    Sustaining Diamond Program Partner

    F. Hoffmann-La Roche, Ltd.

    Sustaining Platinum Program Partner

    AbbVie, Inc.

    Biogen

    MedImmune, a member of the AstraZeneca Group

    Sustaining Gold Program Partner

    Novo Nordisk A/S

    Sustaining Silver Program Partner

    Pfizer, Inc.

    Gold Program Partners

    Agilent Technologies

    Amgen Inc.

    Bill & Melinda Gates Foundation

    Celgene Corporation

    Silver Program Partners

    Baxalta

    BioMarin Pharmaceutical, Inc.

    Bristol-Myers Squibb Company

  • Silver Program Partners - Continued

    Catalent Pharma Solutions

    Eli Lilly and Company

    ProteinSimple

    SCIEX

    Waters Corporation

    Bronze Program Partners

    Bruker Daltonics, Inc.

    Genzyme Corporation, a Sanofi Company

    GlaxoSmithKline

    Friend of CASSS Program Partners

    Bayer Healthcare, LLC

    Janssen Pharmaceutical, Research & Development, LLC

    Seattle Genetics, Inc.

  • Exhibitor Partners

    AAPS

    ABC Laboratories, Inc.

    Agilent Technologies

    Avid Bioservices, Inc.

    BioPhia Consulting, inc.

    Bio-Rad Laboratories, Inc.

    BioReliance Corporation

    Bruker Daltronics, Inc.

    Caprion

    Catalent Pharma Solutions

    Covance, Inc.

    Eurofins Lancaster Labs

    Hunter Lab

    Genedata

    Genovis AB

    KBI Biopharma Inc.

    NanoImaging Services

    New England Biolabs, Inc

    PPD

    Protein Metrics, Inc.

    ProteinSimple

    ProZyme, Inc.

    SCIEX

    SGS Life Science Services

    Thermo Fisher Scientific

    Waters Corporation

    Wyatt Technology Corporation

    Media Partners The Scientific Organizing Committee gratefully acknowledges the following

    media for their promotional consideration of WCBP 2016

    American Laboratory

    American Pharmaceutical Review

    Bio Process International

    BioProcessing Journal

    Bio Tech International

    Genetic Engineering and Biotechnology News

    IPQ Publications

    LCGC North America

    Pharmaceutical Outsourcing

    RSC Advances

    The Analytical Scientist

    The Medicine Maker

    The Pathologist

    SeparationsNOW.com

    Technology Networks Limited

  • Acknowledgements Symposium Co-Chairs Workshop Committee Co-Chairs

    Michael Kennedy, CBER, FDA Anissa Cheung, OVRR, FDA

    Juhong Liu, FDA Michele Dougherty, CDER, FDA

    Brian K. Nunnally, Biogen Joe Kutza, MedImmune, a member of the

    AstraZeneca Group

    Shawn Novick, Seattle Genetics, Inc.

    Steering Committee

    John Dougherty, Eli Lilly and Company Christopher Joneckis, CBER, FDA

    Kathy Francissen, Genentech, a Member of the Edwin Moore, University of Illinois

    Roche Group Ilona Reischl, BASG/AGES

    John Frenz, Alnylam Pharmaceuticals, Inc. Marjorie Shapiro, CDER, FDA

    Robert Sitrin, PATH

    Emeritus Members of the Steering Committee

    Robert Cunico, Pacific Bio Labs

    William Hancock, The Barnett Institute, Northeastern University Michael Kunitani, Marin Analytical Consulting

    Thomas Layloff, Supply Chain Management System

    WCBP 2016 Program Committee

    Sid Advant, Kemwell Biopharma

    Vince Anicetti, Coherus Biosciences

    Mehrshid Alai-Safar, Baxalta, Inc.

    Kris Antonsen, BioMarin Pharmaceutical, Inc.

    Laura Bass, Bristol-Myers Squibb

    Cheryl Blasie, Bristol-Myers Squibb

    Marcus Blȕmel, Novartis Pharma AG

    Andrew Chang, Novo Nordisk, Inc.

    Xiao-Ping Dai, Celgene Corporation

    Bharat Dixit, Genocea Biosciences, Inc.

    John (JR) Dobbins, Eli Lilly and Company

    Roman Drews, LFB-USA

    Julia Edwards, Biogen Idec

    William Egan, GlaxoSmithKline

    Elizabeth Fowler, CMC Consulting Services

    Michelle Frazier, AbbVie, Inc.

    Rajesh K. Gupta, Biologics Quality &

    Regulatory Consultants, LLC

    Reed Harris, Genentech, a Member of the

    Roche Group

    John Hennessey, NovaDigm Therapeutics, Inc.

    Ping Hu, Janssen Pharmaceutical R & D, Inc.

    Stephen Hadley, Bill & Melinda Gates

    Foundation

    David Lee, AbbVie, Inc.

    Karen Lee, Sanofi Global Biotherapeutics

    Kathy Lee, Eli Lilly and Company

    William Matousek, Regeneron Pharmaceuticals,

    Inc.

    Jamie Moore, Genentech, a Member of the Roche

    Group

    Shawn Novick, Seattle Genetics, Inc.

    Joann M. Parker, Pfizer, Inc.

    Stefanie Pluschkell, Pfizer, Inc.

    Lesley Redfern, Abbvie Inc.

    Roberto Rodriguez, Bristol-Myers Squibb

    Mark Schenerman, MedImmune, a member of the

    AstraZeneca Group

    Sally Seaver, Seaver Associates, LLC

    Emily Shacter, Think FDA

    Zahra Shahrokh, STC Biologics, Inc.

    Joseph Siemiatkoski, BioProcess Technology

    Consultants, Inc

    Brian Silvey, Baxalta, Inc.

    Dan Some, Wyatt Technology Corporation

    Arne Staby, Novo Nordisk A/S

    Lisa Stephenson, MedImmune, a member of the

    AstraZeneca Group

  • Chi-Ting Huang, CTH Analytical Consulting

    Peter Johnson, 3M Drug Delivery Systems

    Maura Kibbey, United States Pharmacopeial

    Convention (USP)

    Carol Krantz, ProNAi Therapeutics, Inc.

    Bob Kuhn, Amgen Inc.

    Annie Sturgess, Bristol-Myers Squibb

    Garry Takle, Merck, Sharp and Dohme

    Michael Washabaugh, MedImmune, a member of

    the AstraZeneca Group

    Ziping Wei, Novavax, Inc.

    Yuan Xu, Merck Research Labs

    Heidi Zhang, Genentech, a Member of the Roche

    Group

  • CASSS WCBP 2016 Student Travel Grants

    CASSS is pleased to provide a limited number of student travel grants for PhD students and post-

    docs who present applicable posters at the 20th Symposium on the Interface of Regulatory and

    Analytical Sciences for Biotechnology Health Products (WCBP 2016). PhD students or post-

    doctoral fellows conducting research at academia throughout the world are eligible.

    Requirements are:

    Present a poster on a WCBP 2015 topic

    Proof of studentship/post-doc status

    Recommendation from the supervisor/advisor

    An abstract submission

    A CV for the candidate

    This year’s winners include:

    Development of a Customizable Microwell Array for Controlled Study of Heterogeneous

    Cell Populations

    Natasha Arora and Marianna Sofman

    Massachusetts Institute of Technology (MIT), Boston, MA USA

    Insulin: A Case Study for the Global Supply of a Protein Pharmaceutical

    Raeann Dalton

    The Barnett Institute, Northeastern University, Boston, MA USA

    Rapid 3D Extrusion of Synthetic Tumor Vascular Microenvironments

    Joshua M. Grolman

    University of Illinois at Urbana-Champaign

    Procainamide Labelling as Part of a Flexible Glycoprofiling System for Monitoring of Gal-

    α1-3Gal Related Glycosylation Critical Quality Attributes (GCQAs) of Monoclonal

    Antibody (mAb) Therapeutics Throughout the Product Life Cycle.

    Radoslaw Kozak

    Demasking of Endotoxin in a Drug Product

    Johannes Reich

    University of Regensburg, Regensburg, Germany

    Biopharmaceutical Stability Studies on Therapeutic Proteins with off-line Tandem Mass

    Spectrometry and Real Time Raman Techniques

    Di Wu

    The Barnett Institute, Northeastern University, Boston, MA USA

  • WCBP 2016 Scientific Program Summary

    TUESDAY, JANUARY 26, 2016

    07:00 – 17:00 Registration in the Senate Room

    07:00 – 08:00 Continental Breakfast in the East/State Rooms

    Sponsored by ProteinSimple

    08:00 – 08:15 CASSS Welcome and Introductory Comments

    08:15 – 08:35 5th Annual William S. Hancock Award Announcement in the Grand

    Ballroom

    Sponsored by CASSS and Presented by Wassim Nashabeh, F.

    Hoffmann-La Roche, Ltd.

    08:35 – 08:45 WCBP Welcome and Introductory Comments

    Brian K. Nunnally, Biogen

    08:45 – 09:00 Tribute to Past Chairs of WCBP

    09:00 – 09:45 Keynote Speaker – Gerd Binnig, Definiens AG

    09:50 – 10:50

    Opening Regulatory Panel with the FDA

    Panel Discussion in the Grand Ballroom

    Session Chair: Brian K. Nunnally, Biogen

    Moderator: Stefanie Pluschkell, Pfizer, Inc.

    Panel Members:

    Ashley Boam, Acting Director, CDER/OPQ/OPPQ, FDA, USA

    David Deloski, CDER, FDA USA

    Christopher Joneckis, Associate Director, CBER,, FDA, USA

    Steven Kozlowski, Director, CDER, FDA, USA

    10:50 – 11:15 AM Break – Visit the Exhibits in the East/State Rooms and the Posters in

    the Promenade Room

  • TUESDAY, JANUARY 26, 2016 - Continued

    One Global Specification

    Plenary Session in the Grand Ballroom

    Session Chairs: Neha Frantz, Biogen and Annie Sturgess, Bristol-Myers Scribb

    11:15 – 11:40 Right Sizing Release and Stability Testing, Agency Responses and

    Future Directions

    Barry Cherney, Amgen Inc., Washington, DC USA

    11:40 – 12:05 Chasing the Holy Grail: Global Streamlined Approach to

    Specifications… or Let There Be Light!

    Diane Wilkinson, Biogen, Ltd,, Berkshire, United Kingdom

    12:05 – 12:30 PMDA Perspective on Specifications for Biotechnological Products

    Yasuhiro Kishioka, Pharmaceuticals and Medical Devices Agency

    (PMDA), Japan

    12:30 – 12:45 Panel Discussion - Questions and Answers

    12:45 – 14:15 Lunch Break - Participants on their own

    13:00 – 14:00 Technical Seminars

    Deploying LC/MS Technologies Beyond the Realm of Biotherapeutic Characterization

    Sponsored by Waters Corporation Chinese Room

    NOTE: Lunch is provided for first 100 attendees

    N-Glycan Analysis: From High-Throughput Screening to In-Depth Characterization

    Sponsored by ProZyme, Inc District Room

    NOTE: Lunch is provided for first 100 attendees

    Sponsored by Catalent Pharma Services Palm Court Room

    NOTE: Lunch is provided for first 100 attendees

  • TUESDAY, JANUARY 26, 2016 - Continued

    Pay Now or Pay Later: Diverse Process Development Pathways that Ultimately Support

    Commercialization

    Plenary Session in the Grand Ballroom

    Session Chairs: Xiao-Ping Dai, Celgene and Jamie Moore, Genentech, a Member of the

    Roche Group

    14:15 – 14:40 A Regulatory Perspective on Product Development

    Laurie Graham, CDER, FDA, Silver Spring, MD USA

    14:40 – 15:05 Evaluation of Production from Cell Pools as an Enabling Technology

    for Rapid Advancement of Biologics

    Trent Munro, Amgen, Inc., Thousand Oaks, CA USA

    15:05 – 15:30 Balancing Speed Versus Risk—When to Make Investment Choices in

    Development Projects.

    John Joly, Genentech, a Member of the Roche Group, South San

    Francisco, CA USA

    15:30 – 15:45 Panel Discussion - Questions and Answers

    15:45 – 16:00 Transition Time and PM break for Roundtable Session

    16:00 – 16:45 Roundtable Session:

    Select Your Table Topic in One of Four Rooms

    Chinese Room

    District Room

    Exhibitor Hall (East/State Rooms)

    Palm Court Room

    Please refer to table topics listing in the back of this program book.

    Table seats are on a First Come, First Serve Basis

    16:45 – 17:00 Transition Time

  • 17:00 – 18:15 Workshop Session 1

    Regulatory and Compendial - Combination Products

    Rakhi Dalal, CDER, FDA, Donna French, Genentech, a Member of the Roche Group,

    Steve Hertz, CDER, FDA, Ed Patton, CBER, FDA, Michael Soberg Christensen, Novo

    Nordisk AG, John Weiner, OCP, FDA

    Pay Now or Pay Later: Prioritizing CMC Development Activities in Early Development

    Christopher Downey, CDER, FDA, Catherine Eakin, Seattle Genetics, Inc., Michelle Frazier,

    AbbVie, Inc., Sara Gagneten, CBER, FDA

    Regulators’ Perspectives on Trends in the Regulation of Biopharmaceutical Products in

    Europe and Asia

    Co-chairs: Kathy Francissen, Genentech, a Member of the Roche Group, Anthony Ridgway,

    Health Canada, – Presentations By: Peter Richardson, European Medicines Agency (EMA),

    United Kingdom, Daisaku Sato, Pharmaceuticals and Medical Deveices Agency (PMDA),

    Japan, and Anis Talib, Malaysian Health Authority

    Emerging Trends for Higher Order Structure Characterization in Biopharmaceutical

    Development

    Frances Namuswe, CDER, FDA, Anders Dybdal Nielsen, Novo Nordisk, Andrey Sarafanov,

    CBER, FDA, William Weiss, Eli Lilly and Company

    18:15 – 19:15 Poster Session I in the Cabinet Room

    19:15 – 19:30 Transportation will be provided at 19:15

    19:30 – 22:30 Welcome Reception at the National Archives Museum

  • WEDNESDAY, JANUARY 27, 2016

    07:00 – 17:00 Registration in the Senate Room

    07:00 – 08:40 Continental Breakfast in the East/State Rooms

    08:30 – 08:40 Announcements by Brian K. Nunnally, Biogen

    Implications of Product-Specific Monographs for Biotherapeutic Products

    Parallel Session in the Grand Ballroom

    Session Chairs: John Dougherty, Eli Lilly and Company and Tina Morris, USP

    08:40 – 09:05 The Role of European Pharmacopoeia Monographs in Setting Quality

    Standards for Biotherapeutic Products

    Emmanuelle Charton, EDQM, Council of Europe, Strasbourg, France

    09:05 – 09:30 Implications of Product-Specific Monographs for Biotherapeutic

    Products

    Anthony Mire-Sluis, Amgen, Inc., Thousand Oaks, CA USA

    09:30 – 09:55 The Role of Product-Specific Monographs in Biosimilar Product

    Development

    Xiaoyu Chen, Hospira, a Pfizer Company, Lake Forest, IL USA

    09:55 – 10:10 Panel Discussion - Questions and Answers

    Analytical Control Strategy of Vaccine Products

    Parallel Session in the District Ballroom

    Session Chairs: Arifa Khan, CBER, FDA and Ziping Wei, Novavax

    08:40 – 09:05 Analytic Control Strategies of Vaccine Products During Product

    Development

    Freyja Williams, CBER, FDA, Silver Spring, MD USA

    09:05 – 09:30 GARDASIL®9 Control Strategy Evolution During Late-Stage

    Development and Beyond

    Jennifer L. Dashnau, Merck & Co, Inc., West Point, PA USA

    09:30 – 09:55 Definition of Analytical Control Strategy for Robust Vaccine

    Development

    Christina Campa, GlaxoSmithKline Vaccines, Siena, Italy

    09:55 – 10:10 Panel Discussion - Questions and Answers

  • WEDNESDAY, JANUARY 27, 2016 - Continued

    10:10 – 11:00 AM Break – Sponsored by Pall ForteBio, LLC

    Visit the Exhibits in the East/State Rooms and the Posters in the Cabinet

    Room, or attend one of the Technical Seminars

    10:20 – 10:50 Technical Seminars

    Advances in Analytical Characterization of Biotherapeutics: Powerful Data Processing

    Software Leveraging High-Resolution Accurate Mass data and High-Throughput, High-

    Resolution N-glycan Analysis using Multi Capillary Electrophoresis

    Sponsored by Thermo Fisher Scientific Chinese Room

    At-line and off: The Light Scattering Toolkit for Essential Biophysical Characterization

    Sponsored by Wyatt Technology Corporation Palm Court Room

    11:00 – 12:15 Workshop Session 2

    Product Quality Attributes, Risk Assessment and Control Strategies

    Amin Khan, GlaxoSmithKline, Robin Levis, CBER, FDA, Pat McGeehan, MedImmune, A

    member of the AstraZeneca Group, Leslie Rivera-Rosado, CDER, FDA, Joseph Siemiatkoski, BioProcess Technology Consultants, Inc

    How to Succeed with Breakthrough

    Kimberly May, Merck and Company, Mikhail Ovanesov, CBER, FDA, Emily Shacter, Think

    FDA, Joanna Zhou, CDER, FDA

    Global Regulators Focus Americas

    Gerald DiDonato, Bristol-Myers Squibb, Thomas Schreitmueller, F. Hoffmann-La Roche, Ltd.

    Analytical Control Strategy for Vaccine Products

    Steven Rubin, CBER, FDA, Garry Tackle, Merck, Sharp and Dohme, Michael Washabaugh,

    MedImmune, A member of the AstraZeneca Group, Freyja Williams, CBER, FDA

    12:15 – 14:00 Lunch Break – Participants on their own

    https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=8&cad=rja&uact=8&ved=0CDMQFjAHahUKEwjFwO-NzJ3IAhXGMogKHcmvDRk&url=http%3A%2F%2Fus.gsk.com%2Fen-us%2Fcareers%2F&usg=AFQjCNGqOOzYXMMYwvll4NfAPVBkxgSwLQ&sig2=HP8QMaHjmo83xHITknTVww

  • WEDNESDAY, JANUARY 27, 2016 - Continued

    12:45 – 13:45 Technical Seminars

    Higher Order mAb Aggregate Analysis Using New Innovative Size Exclusion

    Chromatography (SEC) Technology and

    Simplifying 2D-LC, 2D-LC/MS Automated Workflows

    Fast and Efficient Multi-Dimension LC & LC/MS Method Development

    Sponsored by Agilent Technologies Chinese Room

    NOTE: Lunch is provided for first 100 attendees

    Advancing the Intact mAb and mAb Subunit Mass Spectrometry Profiling Workflows

    with Improved Fragmentation Strategies on Ultrahigh Resolution QTOF Sponsored by Bruker Daltonics, Inc. District Room

    NOTE: Lunch is provided for first 100 attendees

    Breakthrough Applications to Expand and Speed Biopharmaceutical Characterization Sponsored by SCIEX Palm Court Room

    NOTE: Lunch is provided for first 100 attendees

    Facing the Challenges of Drug Product and Device Development & Manufacturing

    Plenary Session in the Grand Ballroom

    Session Chairs: Shan Jiang, Seattle Genetics and Margaret Speed Ricci, Amgen Inc.

    14:00 – 14:25 An Integrated, Science and Risk based Approach to the Development

    and Control of Biotechnology Drug Product

    Anthony Mire-Sluis, Amgen Inc., Thousand Oaks, CA USA

    14:25 – 14:50

    Donna French, Genentech, a Member of the Roche Group, South San

    Francisco, CA USA

    14:50 – 15:15 Aseptic Processing of Biological Products: Current Regulatory Issues

    Patricia Hughes, CDER, FDA, Silver Spring, MD USA

    15:15 – 15:30 Panel Discussion - Questions and Answers

    15:30 – 15:45 PM Break and Transition Time

    15:45 – 16:30 Round Table Session II: Select Your Table Topic in One of Three

    Rooms

    Chinese Room

  • District Room

    Exhibitor Hall (East/State Room)

    Palm Court Room

    Please refer to table topics listing in the back of this program book.

    Table seats are on a First Come, First Serve Basis

    16:30 – 16:45 Transition Time

    16:45 – 17:45 Poster Session II in the Cabinet Room

    17:45 – 19:15 Exhibit Reception in the East/State Rooms

  • THURSDAY, JANUARY 28, 2016

    07:00 – 17:00 Registration in the Senate Room

    07:00 – 08:45 Continental Breakfast in the East/State Rooms

    08:30 – 08:45 Announcements by Brian K. Nunnally, Biogen

    Biosimilars Parallel Session in the Grand Ballroom

    Session Chairs: Martin Schiestl, Sandoz and Marjorie Shapiro, CDER FDA

    08:45 – 09:10 Recent Trends in the Evaluation of Analytical Biosimilarity

    Thomas Stangler, Sandoz GmbH, Kundl, Austria

    09:10 – 09:35

    Maria-Teresa Gutierrez-Lugo, CDER, FDA, Silver Spring, MD USA

    09:35 – 10:00 Similarity Assessment of Biosimilars. The Past, Present and Future

    State James Anderson, Momenta Pharmaceuticals, Inc. Cambridge, MA USA

    10:00 – 10:15 Panel Discussion - Questions and Answers

    Comparability for Blood Products – How Did We Get Here and Where Are We Going?

    Parallel Session in the District Ballroom

    Session Chairs: Andrew Chang, Novo Nordisk and Timothy Lee, CBER, FDA

    08:45 – 09:10 Twenty Years of Comparability, Where Are We Now?

    Nancy Kirschbaum, CBER, FDA, Silver Spring, MD USA

    09:10 – 09:35 The Development of Novel Modified Recombinant Blood Factors with

    Extended Half-lives in vivo: A CMC Perspective

    Stephen Raso, Biogen, Cambridge, MA USA

    09:35 – 10:00 How to Mitigate Issues with Potency Assignment Assays for Clotting

    Factor Products - Recent Examples from Baxalta

    Peter Turecek, Baxalta GmbH, Vienna, Austria USA

    10:00 – 10:15 Panel Discussion - Questions and Answers

    10:15 – 11:15 AM Break – Visit the Exhibits in the East/State Rooms and Posters in

    the Cabinet Room

  • THURSDAY, JANUARY 28, 2016 - Continued

    10:30 – 11:00 Technical Seminars

    Using Micro Flow Imaging as a Stability Indicating Method

    Sponsored by Eurofins Lancaster Laboratories Chinese Room

    Structural and Functional Biosimilarity – Concepts and Technical Considerations

    Sponsored by SGS Life Science Service Palm Court Room

    11:00 – 11:15 Transition Time

    11:15 – 12:30 Workshop Session 3

    Stability: Strategy and Expectations for Biotech/Biological Products

    Bazarragchaa Damdinsuren, CDER, FDA, Annick Gervais, UCB, Philip Krause, CBER, FDA,

    Patsy Lewis, Seattle Genetics, Donnie Pullman, Biogen

    Steps Towards Developing a CASSS Global Health Initiative

    Workshop presented through a collaboration between MIT/CBI and CASSS

    John Frenz, Alnylam Pharmaceuticals, Stacy Springs and James Leung, Center for Biomedical Innovation, Massachusetts Institute of Technology, William Hancock, The Barnett

    Institute, Northeastern University

    Advanced Process Control Opportunities for Biologics

    Jay Higgins, Amgen, Inc., Richard Ledwidge, CDER, FDA, Hailun Ma, CBER, FDA,

    Arne Staby, Novo Nordisk

    Facing the Challenge of Drug Product Manufacturing, Validation, and Comparability

    Strategies

    Hung-Wei Chih, Genentech, a Member of the Roche Group, Emily Dubis, Biogen, Chikako

    Torigoe, CDER, FDA, Nicole Trudel, CBER, FDA

    12:30 – 13:45 Hosted Lunch Break in the East/State Rooms

  • THURSDAY, JANUARY 28, 2016 - Continued

    13:45 – 15:00 Workshop Session 4

    Endotoxin Testing: Not Always as Easy as it Seems

    Michael DeFelippis, Eli Lilly and Company, Patricia Hughes, CDER, FDA, Jenny Kenney,

    CBER, FDA, Maura Kibbey, USP, Edwin Moore, University of Illinois

    “Hot Topic” Workshop

    Kathy Lee, Eli Lilly and Company, Nadine Ritter, Global Biotech Experts

    New and Emerging Analytical Technologies that have been driven by the

    Characterization of Biosimilars

    Chi-Ting Huang, CTH Analytical Consulting, Peter Johnson, 3M Drug Delivery, Eva Marszal,

    CBER, FDA, Rachel Novak, CDER, FDA

    Comparability Studies in Blood derived Products and Their Recombinant Analogs

    Mahmood Farshid, OBRR/CBER, FDA, Reed Harris, Genentech, A Member of the Roche

    Group, Alexey Khrenov, OBRR/CBER, FDA, Bryan Silvey, Baxalta, Inc.

    15:00 – 15:30 PM Break in the Promenade Foyer

    15:30 – 15:55 Achieving Product Attribute Control by Implementation of

    Predefined Product Quality Targets and Process Analytical

    Technologies Rohini Deshpande, Amgen, Inc., Thousand Oaks, CA USA

    15:55 – 16:20 Avoiding Facility Obsolescence: Immortal Facilities for Increasing

    Drug Diversity

    Parrish Galliher, Xcellerex Inc. GE Healthcare Life Sciences,

    Marlborough, MA USA

    16:20 – 16:45 Oxygen Uptake as a Virtual Cell Culture Probe

    Steven Rose, MedImmune, A member of the AstraZeneca Group,

    Gaithersburg, MD USA

    16:45 – 17:00 Panel Discussion - Questions and Answers

    17:00 – 17:15 Closing Comments and Invitation to WCBP 2017

    Joseph Kutza, MedImmune, A member of the AstraZeneca Group,

    Next Big Thing in Biologics Processing and Controls

    Plenary Session in the Grand Ballroom

    Session Chairs: Richard Rogers, JUST Biotherapeutics, and Richard Scott Rosenthal,

    MedImmune, A member of the AstraZeneca Group

  • Keynote Speaker

    Gerd Binnig, Definiens AG, München, Germany

    Born in Frankfurt, Germany, Dr. Binnig studied at the J.W. Goethe University in Frankfurt, where

    he received his doctorate degree in 1978. He then immediately joined IBM's Zurich Research

    Laboratory and stayed with IBM till 2002. During this time Dr. Binnig invented and developed

    the Scanning Tunneling Microscope, STM, together with his colleague Dr. Heinrich Rohrer. He

    went on to invent the Atomic Force Microscope, AFM, which he developed together with Calvin

    Quate and Christoph Gerber during a sabbatical at IBM Almaden Research Center (1985/86) and

    a guest professorship at Stanford University (1985-88). Additionally, he opened and headed a

    small IBM research group from 1987 to 1995 within the University of Munich, from which he

    received an honorary professorship.

    Through both techniques, STM and AFM, atoms on the surface of matter are imaged and

    manipulated so that features of single atoms, such as electronic states (STM) and interaction forces

    (AFM), can be measured. The potential of investigating and manipulating matter on the atomic

    scale started the new discipline of nanotechnology. In addition to receiving numerous awards and

    honors, Dr. Binnig was awarded the Nobel Prize in Physics together with his colleague Dr.

    Heinrich Rohrer for the invention of the STM.

    In 1995, Dr. Binnig together with the journalist Dieter Herold founded a small research group,

    which was the precursor of Definiens. In 2000 he founded the company Definiens by bringing in

    investors. With his team at Definiens he developed the Cognition Network Technology, CNT, to

    automatically understand complex data. This technique was initially applied to image analysis

    which uniquely enabled Definiens software to analyze large numbers of images automatically, just

    like the human eye and brain are capable of doing. Later, CNT was extended to the automated

    analysis of data tables derived from the analysis and rich quantification of tissue images, enabling

    the novel field of Tissue Phenomics.

  • Tissue Phenomics – Discovering Spatial Cell Patterns to Predict Drug Response in

    Immunotherapy

    Gerd Binnig, Definiens AG, München, Germany

    Immunotherapy is an exciting and fast emerging field in oncology enabling highly effective

    treatments of cancer patients. Selecting patients, however, for a specific cancer therapy is not

    trivial. For several reasons this is in particular true for immune mediated therapy as the number of

    different treatments will increase drastically in the near future. This is mainly due to the large

    amount of potential combination therapies in this field and it has to be decided which of those

    treatments is best for a specific patient at what stage of his disease. Tissue Phenomics represents a

    novel technique to discover relevant patterns in tissue slides that predict clinical outcome. For

    immunotherapy the characterization of the tumor with its defense mechanisms as well as the state

    and the configuration of the immune cells are most important. Especially the interaction of both

    can be evaluated most precisely through tissue Phenomics. First results for predicting drug

    response are shown for anti-PDL1 NSCLC therapy and anti-PDL1/CTLA4 combination therapy.

    NOTES:

  • Plenary Session Abstracts

    2016 WCBP Opening Regulatory Panel with FDA Session Chair: Brian K. Nunnally, Biogen, Research Triangle Park, NC

    Moderator: Stephanie Pluschkell, Pfizer, Inc., Groton, CT

    Panel Members:

    Ashley Boam OPQ/OPPQ, FDA, USA

    David Deloski, CDER, FDA USA

    Christopher Joneckis, CBER, FDA, USA

    Steven Kozlowski, CDER, FDA, USA

    This year’s opening regulatory panelists represent leading members of FDA who have been invited

    to have an open conversation at WCBP 2016 to discuss key developments and focus areas for the

    FDA that can have substantial impact on the regulatory development pathways of biologics from

    the very early stages of clinical evaluation through license application and market entry. Topics

    will include (1) the reorganization that has established the Office of Pharmaceutical Quality (OPQ)

    and its go-forward imperatives; (2) recent experiences and new perspectives on the acceleration of

    patient access to highly innovative breakthrough biotherapeutic medicines and vaccines, and any

    potential hurdles in this effort; and (3) insights and key learnings from the progression of the

    biosimilar development pathway in the US to date.

    Regarding the first theme, the Office of Pharmaceutical Quality (OPQ) was officially launched in

    January 2015, after having first been announced by longtime Center for Drug Evaluation and

    Research (CDER) director Janet Woodcock in September 2012. "Quality is the underpinning of

    everything we do, and it is imperative that we have a drug quality program as robust as those

    programs we presently have for drug efficacy and drug safety," said Woodcock in a 2012 memo

    to FDA staff. The conversation with the panelists will include details on the organizational changes

    and how they have impacted or are intended to impact both pre- and post-approval quality

    oversight, the speed of reviews and decision making, and the industry’s implementation of

    innovative, superior manufacturing technologies along the entire lifecycle of a product. In addition,

    the ability of the new OPQ to manage the reality of increasingly complex global supply chains for

    the manufacture of medicines and vaccines and the sourcing of raw materials will also be

    discussed.

    On the topic of accelerated development pathways, the dialogue will include an update on FDA’s

    recent experience with its programs that are intended to expedite patients’ access to critical

    medicines, including therapies designated as “breakthrough”. The focus will center on the

    challenges of expedited clinical development on the sponsors’ CMC organizations and any

    recommendations that FDA can make on how to prioritize and streamline the elements of

    regulatory CMC requirements either before or after market entry (may also include specifics for

    combination products, devices). Furthermore, a discussion of a variety of issues that can hinder

    innovation and speed of development will also take place, including any obstacles to science- and

    risk-driven decisions on manufacturing process and quality testing strategies (e.g. the role of

    product-specific monographs for biotherapeutics).

  • For the discussion on biosimilars, we look forward to FDA’s insights into their growing experience

    interacting with sponsors of biosimilar development programs, pre-approval expectations to

    demonstrate biosimilarity, and the evolution of FDA’s biosimilar guidelines. The FDA’s

    perspective on their requirements for biosimilar market entry relative to other geographical regions

    such as the EU or Canada, biosimilar naming (e.g. relative to WHO proposals) and product labeling

    will also be covered.

    NOTES:

  • Chasing the Holy Grail: A Global Approach to Specifications:

    Session Chairs: Neha Frantz, Biogen and Annie Sturgess, Bristol-Myers Squibb

    Specifications ranges for biotherapeutics have traditionally been defended based on process

    capability and statistical analysis of manufacturing experience. As the industry implements the

    principles of quality by design (QbD) both in bioprocess and assay development as well as and

    “fit-for-purpose” control strategies, we find ourselves at odds between increased product and

    process understanding and antiquated arguments for defending the tests and acceptance criteria

    that define the specifications that support commercial registration and life-cycle management of

    products globally.

    Building on the WCBP themes since 2013 with the mindset of global regulatory convergence, and

    driven by the increasing complexity of the biotherapeutics commercialized world-wide, this

    plenary session will explore through case studies the industry’s desire to move to a scientifically-

    sound, patient-centered and risk-based approach to developing and defending specifications, with

    the goal of ensuring and maintaining the integrity of global supply.

    NOTES:

  • Right Sizing Release and Stability Testing, Agency Responses and Future Directions

    Barry Cherney, Amgen Inc., Washington, DC USA

    Amgen has performed a holistic review of our integrated control strategy utilizing QbD principles

    including scientific understanding and risk based assessments to create a streamlined, focused

    testing program for our products that eliminates non-value added testing while maintaining or

    enhancing our ability to ensure product quality with a high degree of confidence. While the initial

    implementation of this program was described during WCBP 2015, we have now expanded the

    program to include multiple products and regulatory jurisdictions. This presentation will provide

    an update on our progress, using real life examples and will describe the feedback we have received

    from the various regulatory authorities and the challenges we face in fully implementing our

    strategy. In addition to the right size testing initiative, Amgen is leveraging advances in mass

    spectrometry that are capable of replacing many of the methods currently used in establishing

    specifications. The utility of this method and the regulatory implications and challenges will be

    discussed along the role a robust pharmaceutical quality system can play in defining a science and

    risked based control strategy that allows manufactures increased flexibility to make appropriate

    decisions.

    NOTES:

  • Chasing the Holy Grail: Global Streamlined Approach to Specifications… or Let There Be

    Light!

    Diane Wilkinson, Biogen, Ltd., Berkshire, United Kingdom

    A key component of Pharmaceutical Product Quality controls are the specifications developed

    based on science and agreed prior to and during review and approval with worldwide Regulatory

    Agencies. Traditionally these are based on data from batches tested prior to application, in clinical

    efficacy and safety assessment, with statistical analysis applied. Over the last few years the

    Industry has implemented aspects of the principles of quality by design (QbD) in bioprocess and

    in assay development and developed “fit-for-purpose” control strategies, as part of Quality

    Management systems. So we therefore find ourselves in a situation where we need to take a fresh

    approach to defining and agreeing specifications for development, commercialization and life-

    cycle management of products and continue to strive for globalization of the approaches.

    In this presentation we explore the current activities taking place in Europe and globally to move

    forward with fresh approaches, via identification of regulatory pathways to expedite CMC

    development and in new guidance development. The concepts of PRIME and Adaptive Pathways

    and their place in defining agreement to specifications will be reviewed.

    From a guidance perspective, while acknowledging that the concepts in ICH Q8, Q9, Q10 and Q11

    provide opportunities for a more science- and risk-based approach for assessing changes across

    product lifecycle, including to specifications, there are still opportunities to improve and expand

    how specifications can be developed and controlled. We will therefore also look at the potential

    impact of developing new guidance e.g. ICH Q12, where a framework to facilitate the management

    of post-approval CMC changes in a more predictable and efficient manner across the product

    lifecycle is being developed. This will include exploring approaches for improved post-approval

    ‘operational flexibility’, through alignment on the necessary information and level of detail in the

    dossier and its impact on change management and regulatory reporting, for established conditions.

    Concepts of improved utilization of post-approval change management plans and comparability

    protocols will also be discussed. It is hoped from progression of this guidance, that the benefits of

    global development and changes to global specifications will be improved on its adoption, whilst

    still ensuring patient safety, efficacy and quality. We will look at the role played by Industry in

    this, in partnering with Trade Associations, and Regulatory Agencies and bodies, to achieve our

    goal, of scientifically-sound, patient-centered and risk-based approaches to develop and defend

    specifications and move closer to our holy grail of global specifications.

    NOTES:

  • PMDA Perspective on Specifications for Biotechnological Products

    Yasuhiro Kishioka, Pharmaceuticals and Medical Devices Agency (PMDA), Japan

    Since insulin was since the first recombinant insulin was approved in 1985, more than 100

    biotechnological products have been approved in Japan. Based on this experience, this presentation

    will give PMDA perspective on specifications for biotechnological products.

    NOTES:

  • Pay Now or Pay Later: Diverse Process Development Pathways that Ultimately Support

    Commercialization

    Session Chairs: Jamie Moore, Genentech, a Member of the Roche Group, and Xiao-Ping

    Dai, Celgene

    This plenary session will highlight two distinct aspects of CMC development: CMC activities

    completed proactively in early development that can expedite late stage development; accelerated

    early phase CMC development to support fast to FIH and proof of concept.

    It will consist of case studies examining successful technical development approaches such as

    implementation of molecular/developability assessment, developing comprehensive host cell

    protein (HCP) assays, sequence variant analysis, establishing clonality; as well as strategies to

    speed development and delivery for FIH.

    NOTES:

  • A Regulatory Perspective on Product Development

    Laurie Graham, CDER, FDA, Silver Spring, MD USA

    The use of 21st century pharmaceutical principles, such as those described in the International

    Conference on Harmonization (ICH) Guidelines Q8-Q11, is expected to result in increased

    product and process understanding leading to enhanced, risk based control strategies. During early

    product development, the use of these principles can strengthen and provide flexibility to the CMC

    development strategy. For example, as described in ICH Q9, an effective quality risk management

    approach is expected to not only provide a proactive means to identify and control potential quality

    issues, but also to facilitate better and more informed development decisions. Ms. Graham will

    give a regulatory perspective, with case studies, on how early CMC efforts, in the context of 21st

    century principles, can impact different stages of the product lifecycle.

    NOTES:

  • Evaluation of Production from Cell Pools as an Enabling Technology for Rapid

    Advancement of Biologics

    Trent Munro, Amgen, Inc., Thousand Oaks, CA USA

    With an increasing number of candidate molecules under development, probing the biology as

    quickly as possible is critical for rapidly delivering next generation therapies to patients with unmet

    medical need. During advancement of a biologic to the clinic, cell cloning and subsequent process

    development activities are on the critical path and directly impact the time for clinical introduction

    of a biotherapeutic. Program acceleration is possible if cell pools can be leveraged for early

    material generation and process development activities. To successfully use cell pools during

    development, it is important for cell-pool derived product quality attributes to be comparable to

    those derived from clones. To better understand the relationship between pool and clone derived

    product quality attributes, we compared data across recent programs. Cell pool and clone derived

    material was purified and tested for a number of product quality attributes including expression

    stability, post-translation modifications, and higher order species. Overall, our results indicate

    feasibility of matching product quality attributes between cell pools and subsequently derived

    clones. These findings support the strategic use of cell pools to accelerate the advancement of

    biologics to the clinic.

    NOTES:

  • Balancing Speed Versus Risk—When to Make Investment Choices in Development Projects.

    John Joly, Genentech, a Member of the Roche Group, South San Francisco, CA USA

    In the world of biopharmaceutical development speed to market is critical for bringing innovative

    medicines to patients with significant unmet medical needs. Whether its breakthrough

    designations or priority reviews, the ability to move swiftly through development is prized by

    patients and companies alike. When positive data emerges from early trials, timelines are

    scrutinized and the frequent mantra of “can you go faster?” is heard from many corners. With an

    ever constant focus on development timelines, project teams are often confronted with balancing

    speed versus risk. In this talk I’ll present several vignettes of when to take risks and when to

    further invest time and resources into late stage research and development projects. The choices

    are important ones as only a fraction of the development pipeline will make it to commercial launch

    and in order to efficiently produce successful products, one must decide thoughtfully when to

    invest and when it’s prudent to defer investment to a later stage. I’ll discuss how a rich

    development history with monoclonal antibodies provides platform knowledge that informs a

    development organization of the risks involved with such choices.

    NOTES:

  • Implications of Product-Specific Monographs for Biotherapeutic Products

    Session Chairs: John Dougherty, Eli Lilly and Company and Tina Morris, USP

    Product-specific monographs for biologics have been used in the major pharmacopoeias of the

    world since the early 20th century. As biologics manufacturing, development and quality control

    have changed over time, especially with the introduction of recombinant biotherapeutics into the

    market, development challenges and expectations for compendial standards have also evolved.

    Most recently, the global discussion about quality expectations for biosimilars has added another

    dimension in considering the role of product-specific compendial standards. This session will bring

    together different viewpoints on the value, role, and challenges in the development, applicability

    and use of compendial monographs for modern biotherapeutic medicines. In addition, the session

    will endeavor to establish a conceptual framework for productive dialogue around the use of

    standards in enabling access to quality biotherapeutic medicines for patients worldwide.

    NOTES:

  • The Role of European Pharmacopoeia Monographs in Setting Quality Standards for

    Biotherapeutic Products

    Emmanuelle Charton, EDQM, Council of Europe, Strasbourg, France

    The European Pharmacopoeia (Ph. Eur.) sets up quality standards for medicinal products and their

    constituents that are legally binding in Europe and accepted in countries from all over the world.

    It has always been committed to adapting to a fast developing environment, keeping pace with the

    advancement of scientific technologies and taking care of regulatory needs. These achievements

    apply also to biotherapeutic products, a class of products to which the Ph. Eur. has devoted the

    highest attention over decades. With the appearance of new generation biotherapeutics, new

    challenges have to be solved. The presentation will show, with the support of specific case studies,

    how the Ph. Eur. has overcome the difficulties linked to these products and which challenges

    remain ahead.

    NOTES:

  • Implications of Product-Specific Monographs for Biotherapeutic Products

    Anthony Mire-Sluis, Amgen, Inc., Thousand Oaks, CA USA

    Traditional small molecule product specific pharmacopoeia monographs serve several purposes,

    such as ensuring a consistent approach to quality for innovator and generic products, assessing the

    quality of drug products in commerce, providing specifications that new manufacturers can target,

    monitoring for counterfeit and substandard products as well as monitoring the quality of imported

    drug products. However, there are challenges with creating product monographs for biotechnology

    products since they are inherently complex proteins that generally contain multiple product

    variants. The criticality of such variants differs – some impacting safety or efficacy whilst others

    do not. However, specification tests and their associated limits for some attributes are often set to

    assure consistency of production and may have no impact on safety and efficacy directly. In

    addition, specifications often change during the product lifetime, and thus are only a snapshot in

    time. Setting a single set of limits is not necessarily relevant for example, for terminal

    heterogeneity of a monoclonal antibody product, where variants have been shown to have no

    quality, safety or efficacy impact and can be very process and even site specific. The same applies

    for the glycosylation patterns for biotechnology products where some glycosylation structures are

    impactful while others are not. Therefore, applying attribute limits of the innovator to biosimilars

    may not be meaningful since a biosimilar can have analytical differences that have no clinical

    impact. Other issues apply to the testing and limits for host cell proteins where company/process

    specific test reagents and standards restrict the ability to compare between products. Alternative

    approaches to help assure the quality of biotechnology products have been developed in the form

    of class specific method chapters such as USP for assays for monoclonal antibodies as well

    as chapters that cover best practices for glycosylation and host cell protein testing amongst others.

    NOTES:

  • The Role of Product-Specific Monographs in Biosimilar Product Development

    Xiaoyu Chen, Hospira, a Pfizer Company, Lake Forest, IL USA

    The compendial standards defined in pharmacopoeia product-specific monographs include

    reference materials, testing methodologies and acceptance criteria. These standards are used to

    establish a common set of requirements for product identity, strength, quality, and purity testing,

    and are of great value in the development of small molecule generic products. Biosimilar products,

    however, are regulated based on the totality of the data from comparative analytical, non-clinical,

    and clinical studies for the reference product and proposed biosimilar product. The goal of a

    biosimilar development program is to demonstrate a high degree of analytical similarity between

    the proposed biosimilar product and the reference product for attributes defined as Critical Quality

    Attributes (CQAs). Due to the complexity of protein structures and functions and the

    manufacturing processes used for the production of therapeutic protein products, individual

    manufacturers also often adopt different control strategies based on their own product and process

    knowledge to ensure control of the relevant CQAs. Standardized compendial methods may be

    insufficient to address all possible product-related and process-related impurities for recombinant

    therapeutic proteins produced using different processes and having different formulations. In

    addition, extensive additional characterization above and beyond standard release and stability

    testing using internally developed or compendial methods is required for demonstration of

    biosimilarity. Therefore, the monograph physicochemical and biological tests and associated

    pharmacopoeia reference standards have different roles in the development of biosimilar products

    relative to their roles in the development of generic medicines. In this presentation, the utility and

    challenges of applying product-specific monographs will be discussed in the context of biosimilar

    product development.

    NOTES:

  • Analytical Control Strategy of Vaccine Products

    Session Chairs: Arifa Khan, CBER, FDA and Ziping Wei, Novavax

    This plenary session will discuss how the analytical control strategy evolves over the course of

    vaccine product development. Stage-appropriate and risk-based analytical control strategy needs

    to be developed to meet the product development needs. At the early stages of vaccine product

    development, analytical control strategy is based on limited product understanding and process

    knowledge. As the product moves to late stage development, product understanding and process

    knowledge accumulate and it is expected that an enhanced analytical control strategy is established

    to ensure product quality. As an example, product development for a Phase 1 clinical program,

    where the major concern is patient safety and less product knowledge is gained, would likely have

    a very different analytical control strategy in comparison to one being performed for a Phase 3

    pivotal study, where safety and efficacy are relevant factors and more product knowledge has been

    accumulated. The wide range of vaccine products (which include recombinant proteins, live-

    attenuated viruses, inactivated viruses, polysaccharides, and polysaccharide-protein conjugates)

    present unique aspects and challenges for establishing analytical control strategy. Depending on

    the complexity of vaccine type, it may be challenging to characterize or measure quality attribute

    changes, and define complexity of process and product attributes. This plenary session will give

    examples for how to set control strategy when there is limited product and process knowledge and

    how it evolves as vaccine products move to late stage development.

    NOTES:

  • Analytic Control Strategies of Vaccine Products During Product Development

    Freyja Williams, CBER, FDA, Silver Spring, MD USA

    As products progress through clinical development, chemistry, manufacturing and control

    information must be submitted to the regulatory agency appropriate to the clinical phase of the

    product development. The requirements for specific control testing for each clinical phase cannot

    be predefined but will depend on several factors, including the overall support for safety of the

    product. The regulations applicable to investigative new drugs, and considerations when applying

    them to specific applications will be discussed.

    NOTES:

  • GARDASIL®9 Control Strategy Evolution During Late-Stage Development and Beyond

    Jennifer L. Dashnau, Merck & Co, Inc., West Point, PA USA

    GARDASIL®9 (HPV 9-valent Vaccine, Recombinant), is a nonavalent, recombinant vaccine

    against human papillomavirus – the main cause of cervical, vulvar, vaginal, and anal cancers. This

    second-generation vaccine combines the existing four types found in GARDASIL® (HPV Types

    6, 11, 16, 18), with five additional types (HPV Types 31, 33, 45, 52, 58). HPV vaccines are based

    on virus-like particles (VLP), which are complex structures consisting of approximately 72 L1

    (major capsid protein) pentamers (capsomeres) arranged in an icosahedral formation. Although

    their structure is complex, VLPs are amenable to characterization through a wide range of

    analytical techniques and robust control strategies may be developed. GARDASIL®9 provides an

    opportunity to understand the evolution of analytical control strategy for a well-characterized,

    recombinant vaccine product from late-stage development to launch. Development of the

    additional types was based largely on the first-generation process, thus allowing for leverage of

    existing process and product understanding to inform development of the control strategy – from

    selection of critical quality attributes to justification of specifications. However, challenges

    encountered during late-stage development required modifications be made to some steps of the

    process. For the attributes associated with these steps, more limited data were available, thus

    requiring a different approach for setting control strategy. Finally, as the product enters the launch

    phase, the control strategy continues to be enhanced as increased understanding of process and

    product performance is gained during continued process verification. The presentation will give

    examples for how to set control strategy in these situations.

    NOTES:

  • Definition of Analytical Control Strategy for Robust Vaccine Development

    Christina Campa, GlaxoSmithKline Vaccines, Siena, Italy

    The establishment of a strong Analytical Control Strategy is instrumental to achieve consistent

    manufacturing of safe and efficacious products. For this reason, it is critical to build analytical

    knowledge since early development, leading to a systematic increase of product and process

    understanding during life cycle. This presentation will focus on some key elements allowing the

    realization of such challenging objective:

    Definition of requirements of analytical methods (Analytical Target Profile, ATP), applicable to lot release/ stability testing, characterization/

    comparability testing and process monitoring

    Systematic reliability evaluation of methods for the scope, including considerations on reference standard use and suitability

    Continued refinement of analytical approaches for ATP fulfillment during product life cycle, ensuring application of up-to-date analytical

    technologies

    Implementation of Quality by Design in method development (e.g., identification & ranges of critical variables impacting analytical result),

    with assessment of implications on method qualification/ validation, as

    applicable.

    Case study examples will be provided on vaccine development, demonstrating how the

    implementation of the above- mentioned concepts can be successfully achieved in this field.

    NOTES:

  • Facing the Challenges of Drug Product and Device Development & Manufacturing

    Session Chairs: Margaret Speed Ricci, Amgen Inc. and Shan Jiang, Seattle Genetics

    This plenary session will cover the perspectives of both regulators and industry on challenges in

    drug product and device development and manufacturing for therapeutic biologics. The criticality

    of integrated drug product design considerations across formulation, drug product process,

    container, and device will be discussed. Learnings from the development and commercialization

    of adaptive manufacturing systems and new container and device technologies will be reviewed.

    Establishing comprehensive control strategy and drug product comparability for aseptic

    manufacturing of biologic products will be demonstrated with industry examples. Case studies will

    be shared that illustrate technical considerations in container closure integrity, extractables and

    leachables testing, process validation as well as new regulatory expectations for manufacturing of

    parenteral products.

    NOTES:

  • An Integrated, Science and Risk based Approach to the Development and Control of

    Biotechnology Drug Product

    Anthony Mire-Sluis, Amgen Inc., Thousand Oaks, CA USA

    The development of commercial drug product has to start at the earliest phases of development in

    considering the target product profile (TPP). The TPP should define the intended use of the

    product and consider the dosage form and container and/or device associated with it. One cannot

    ignore the fact that the drug product is derived from drug substance, which is derived from the

    protein produced by a selected production cell clone. Therefore, the TPP should be considered

    when selecting the protein sequence during molecule assessments, since this selection will have a

    huge impact on the nature of the final drug product. Often a first in human drug product will be

    in a different formulation and container than the final commercial product (e.g. frozen in a vial)

    and thus commercial formulation plays a crucial role in defining the final drug product and its

    stability. Therefore, end to end studies to show the ability of product to remain stable at room

    temperature after storage at the recommended storage temperature and/or cycling studies can

    provide the necessary data to support not only excursions during transport, but allow for patients

    to keep product at room temperature for a time before use. The process of formulation and filling

    cannot be ignored either as it can have impact on the quality attributes of the product. For example,

    how the material is pumped through delivery needles (peristaltic versus time/filler pressure) can

    impact subvisible particle load. How drug product is stored during formulation and filling can

    impact bubble formation during filling. Lastly, drug product doesn’t magically appear in the hands

    of a doctor or patient – it has to have a cold chain to get it from the company warehouse to delivery

    to the patient in a way that retains all the desired quality attributes. Control of transport and storage

    at the final destination also plays its part in providing quality drug product to patients. Taken

    together, these concepts create the basis for an integrated control strategy for drug product that can

    utilize risk based approaches for implementation.

    NOTES:

  • Donna French, Genentech, a Member of the Roche Group, South San Francisco, CA USA

    NOTES:

  • Aseptic Processing of Biological Products: Current Regulatory Issues

    Patricia Hughes, CDER, FDA, Silver Spring, MD USA

    An overview of the quality assessment by microbiology product quality reviewers at the

    FDA/CDER of the drug product section of a Biological License Application (BLA) will be

    presented. Examples of recurring application deficiencies and resolutions for BLA approval will

    be presented. The presentation will also discuss the relationship between inspectional findings

    during pre-approval inspection with the application quality assessments for BLA approval.

    NOTES:

  • sEvolving Biosimilar Regulatory Science – Case Studies

    Session Chairs: Martin Schiestl, Sandoz, and Marjorie Shapiro, CDER, FDA

    The regulation of biosimilars is founded on the scientific principles of comparability, which were

    established - with the publications of the FDA Guidance “Demonstration of Comparability of

    Human Biological Products, Including Therapeutic Biotechnology-derived Products” in April

    1996, followed by the ICH Q5E concept paper established in 2002 and the final adoption of ICH

    Q5EComparability of Biotechnological/Biological Products Subject to Changes in Their

    Manufacturing Process in 2004/2005. Prior to this, a biological product was often defined by its

    manufacturing process. Without the acceptance of the concept of “comparability”, along with

    advances in analytical methods, there would be no biosimilars today.

    The comparability concept has also been used for the approval of follow-on-versions of biological

    products regulated in the US for historic reasons as drugs using the 505(b)(2) pathway. One

    example is a Somatropin approved by FDA in 2006 based on comparative quality, non-clinical

    and clinical data. In the same year the same product was also approved as the first biosimilar

    product in the EU using basically the same data package.

    In the EU, EMA issued the first biosimilar guideline already in 2004 and the first products were

    approved in the following years. In the US, the legal basis was created by the Biologics Price

    Competition and Innovation act in 2009. Since then, the regulatory science evolved quickly and

    continues to move forward, for example, new concepts with regard to quality evaluation taking

    into account the clinical relevance of quality attributes, tailored confirmatory clinical studies, and

    the scientific requirements for extrapolation have been developed allowing the science-based and

    efficient development of safe and effective biosimilars. The revised and newly established

    biosimilar guidances in US and EU reflect the current state-of-the-art in regulatory science, while

    remaining open for further innovation in the field. This is important for the next step in the

    evolution: the move from biosimilars to interchangeable biologics.

    This session will illustrate this evolution in regulatory science with case studies of approved

    biosimilar products or those currently in development. Speakers from FDA and Sponsors

    developing biosimilar products will share their experience and provide an outlook to the future.

    NOTES:

  • Recent Trends in the Evaluation of Analytical Biosimilarity

    Thomas Stangler, Sandoz Biopharmaceuticals, GmbH, Tirol, Austria

    The scientific principles of comparability are the foundation for the development of a biosimilar.

    Since the first approval of a biosimilar in the European Union in 2006, analytical technologies,

    development concepts and regulatory sciences have continued to evolve. More and more powerful

    analytical methods, applied to increasing numbers of reference product batches over many years,

    result in larger and larger data sets as basis for the evaluation of analytical biosimilarity. This

    wealth of data offers opportunities and challenges.

    This talk will present recent experiences in Sandoz’ biosimilar development projects and

    regulatory interactions with respect to the evaluation of biosimilarity, especially with respect to

    the application of statistical approaches. Benefits and limitations of statistical approaches will be

    discussed, in addition to how the statistical results fit into the overall evaluation of biosimilarity.

    NOTES:

  • Maria-Teresa Gutierrez-Lugo, CDER, FDA, Silver Spring, MD USA

    NOTES:

  • Similarity Assessment of Biosimilars. The Past, Present and Future State

    James Anderson, Momenta Pharmaceuticals, Inc., Cambridge, MA USA

    This talk will provide historical reference for concepts currently being applied for biosimilars

    followed by an industry perspective on current regulatory guidance for the evaluation of

    physiochemical and biological characterization data (analytical similarity). Challenges associated

    with these assessments – especially early in a development program – will be highlighted.

    A look to the future for establishing analytical similarity will also be presented, specifically the

    application of the “science of comparison” including the incorporation of advanced analytics with

    biological models.

    NOTES:

  • Comparability for Blood Products – How Did We Get Here and Where Are We Going?

    Plenary Co-chairs: Andrew Chang, Novo Nordisk, Inc. and Timothy Lee, CBER, FDA

    This plenary session will address comparability issues related to plasma-derived and recombinant

    blood coagulation factor products. Our speakers will describe how the comparability approach has

    been applied, evolved and leveraged, against a historical backdrop of advancements of

    recombinant technology in product manufacture, improvements in analytical capability and

    changes in regulatory policies. We will set the stage by reviewing the comparability paradigm as

    it is applied throughout the past 20 years since its inception. We will then look at case studies

    illustrating some of the challenges related to specific classes of coagulation factor products, some

    of which are unique to coagulation factors, and others which are relevant to biological products in

    general. In the examples, we hope to cover the following topics:

    • How differences in quality attributes in pre- and post-change materials were reconciled either

    during product development or post-approval

    • How interactions between the manufacturer and regulator helped resolve comparability-related

    issues

    • What are the considerations in the selection of analytical methods for the comparability exercise?

    Are new analytical methods always considered?

    • What are the characteristics that the manufacturer considers to be unique to blood products, and

    those that are shared amongst other biopharmaceuticals, and how that affected the design of the

    comparability exercises? For example, impurities in plasma-derived products could have their own

    biological activities that can either be beneficial or deleterious.

  • Twenty Years of Comparability, Where Are We Now?

    Nancy Kirschbaum, CBER, FDA, Silver Spring, MD USA

    Twenty years ago, FDA published its first guidance document on comparability. Initially, the

    comparability paradigm focused on supporting specified manufacturing changes within a single

    manufacturer by generating relevant analytical, non-clinical and/or clinical PK/PD data, as

    necessary, in a one-time exercise designed to obviate the need to repeat clinical efficacy trials.

    Changes to plasma derivative manufacturing processes were often supported by complementing

    analytical data with non-clinical and/or clinical data in the face of residual uncertainty, despite

    advances in analytical capabilities and manufacturing science. Since then, industry and regulators

    have partnered to implement the life-cycle approach to biopharmaceutical development and there

    is now a legal pathway for applying the comparability paradigm to demonstrating biosimiliarity

    across manufacturers. Moreover, there have been unprecedented advances in analytical science

    and its application to development of cutting-edge in vitro functional tests. This talk will examine

    improvements in analytical capability in the context of changes in regulatory policies to highlight

    the benefits of implementing a risk-based, life-cycle approach to comparability, including

    strategies for leveraging in vitro functional tests that reflect all critical biological functions.

    NOTES:

  • The Development of Novel Modified Recombinant Blood Factors with Extended Half-lives

    in vivo: A CMC Perspective

    Stephen Raso, Biogen, Cambridge, MA USA

    This talk will address not only Biogen’s approach to establishing comparability of biologics

    throughout product development and commercialization, but some of the challenges unique to the

    development and characterization of recombinant blood factor Fc-fusion proteins. Some of these

    additional considerations include the assessment of new functionalities of the modified proteins,

    comparison to existing treatments and alignment with international blood factor standards.

    NOTES:

  • How to Mitigate Issues with Potency Assignment Assays for Clotting Factor Products -

    Recent Examples from Baxalta

    Peter Turecek, Baxalta GmbH, Vienna, Austria

    There are different reasons for measuring clotting factor activities: diagnosis and assessment of

    severity of the clotting factor defect, assessment of the hemostatic status of patients´ post infusion

    samples, determination of recovery, trough levels and pharmacokinetics of concentrates, and

    potency designation by the pharmaceutical manufacturer. These reasons could be subject to

    conflict as the requirements for the assays used for these different purposes might be different.

    Moreover, analytical issues have been reported with modified coagulation factor VIII (FVIII) and

    also factor IX (FIX). There are also geographic differences in the use of certain assays and also

    legal test requirements may differ for one and the same clotting factor.

    In a recent workshop on “Characterization of new clotting factor concentrates (FVIII, FIX) with

    respect to potency assays used for labelling and testing of post infusion samples” the following

    key points were identified (Dodt J, et al. Haemophilia 2015):

    Thorough characterization of new rFVIII and rFIX products, according to International Society of Thrombosis and Hemostasis (ISTH) recommendations, in a variety of potency

    assays against the WHO IS (concentrate and plasma) is important.

    For some modified rFVIII products and for all new rFIX products, one-stage clotting assay methods result in different potency assignments depending on the APTT reagent used.

    For FVIII it was also said that clinicians wish to have one assay that could be used to assign

    potency for product labelling and which correlates with FVIII levels measured by the clinical

    laboratory. A particular concern is that using different assays could lead to discrepant high product

    plasma values measured post-infusion and result in underdosing of patients.

    The potency labeling strategies of two recently approved Baxalta treatments for hemophilia will

    be presented in light of current requirements and demands from patients and physicians.

    NOTES:

  • Back to the Future: The Next Big Thing in Biologics Processing and Controls

    Plenary Co-chairs: Richard Rogers, JUST Biotherapeutics, and Richard Scott

    Rosenthal, MedImmune, A member of the AstraZeneca Group

    The tools, technologies and hardware that enabled the establishment of the biotechnology industry

    continue to evolve and change, and as a result the current state of the industry looks very different

    than it did in its infancy. Much of this evolution occurs as a result of incremental improvements,

    which over time results in greater efficiency, capacity, robustness, utility, etc. Fundamental

    paradigm shifts occur far less frequently, but can result in true step changes in terms of how the

    industry operates. This session seeks to highlight disruptive technologies and processing trends

    with the potential to bring about transformative change to the current state of the industry with

    respect to bioprocessing and product attribute controls.

    NOTES:

  • Achieving Product Attribute Control by Implementation of Predefined Product Quality

    Targets and Process Analytical Technologies

    Rohini Deshpande, Amgen Inc., Thousand Oaks, CA USA

    Quality by Design (QbD) is a systematic approach to development of pharmaceuticals that begins

    with pre-defined objectives, and emphasizes product and process understanding and attribute

    control, based on scientific understanding and quality risk management. This requires an

    understanding of how product, materials, methods, and process variables influence final product

    quality. An integrated product development paradigm starts with a well-defined TPP and QTPP

    to implement attribute control strategies that allow for better real time control of process and

    product. Such an approach, if successfully implemented will permit testing on the manufacturing

    floor while at the same time move the testing paradigm of biologics towards RTRT. In this

    presentation, we will share our progress in application of a QTPP and process analytical

    technologies for achieving attribute control and designing quality into the product.

    NOTES:

  • Avoiding Facility Obsolescence: Immortal Facilities for Increasing Drug Diversity

    Parrish Galliher, Xcellerex Inc. Marlborough, MA USA

    As worldwide annual sales of biologics increase toward the 200 bn USD mark, so has the diversity

    of treatment modalities and drug pipelines increased. Pipeline diversity has grown from

    recombinant hormones and cytokines to monoclonal antibodies (MAb), MAb-toxin conjugates,

    Mab antibody fragments, multivalent MAbs, cell-based and rDNA vaccines, precision cell and

    gene therapies, therapeutic enzymes, biobetters, biosimilars, biofuels and more. Over the decades,

    many manufacturing facilities have been unable to accommodate this increasing diversity and have

    become underutilized, mothballed or obsolete. The presentation will discuss the challenges of

    increasingly diverse pipelines and process flow architectures and how innovations in

    manufacturing facility design and operations can maximize facility utilization and help avoid

    facility obsolescence.

    NOTES:

  • Oxygen Uptake as a Virtual Cell Culture Probe

    Steven Rose, MedImmune, a member of the AstraZeneca Group, Gaithersburg, MD USA

    During the scale-up and tech transfer of cell culture processes, we routinely evaluate whether the

    bioreactors are capable of supporting the required cell culture oxygen demands. We have

    developed a first principle oxygen transfer model to define how best to run the bioreactor to meet

    the oxygen demand. The model enables us to successfully specify larger scale bioreactor operating

    conditions without needing to perform an engineering run. We have found that we can also

    leverage this engineering model as a virtual probe to monitor and control the cell culture process.

    We have shown that with a virtual oxygen uptake rate probe, we can manage the carbon dioxide

    accumulation and predict titer and cell culture growth performance. This provides greater insight

    into the biology and results in consistent control for enhanced process performance. This

    presentation will share the model results and its predictive ability across bench, pilot and

    manufacturing scales.

    NOTES:

  • NEW WCBP 2016 WORKSHOP DESCRIPTIONS

    Expanding the Potential

    Different Workshop topics need to be approached in different ways in order for their full potential

    to be reached. WCBP 2016 will feature three different Workshop formats to enhance discussion

    of key topics of interest.

    1. The Standard Format

    This is the Workshop you usually think of as being associated with WCBP. Industry and

    Regulatory Agency co-chairs focus on generating discussion over the entire period of the

    session. As with all Workshop Formats, audience participation is critical to success.

    2. The Speed-Dating Format

    This isn’t your grandmother’s Workshop! We’ve kicked it up a notch by essentially

    facilitating 4 simultaneous round table sessions on four related Workshop issues. Every

    attendee has the opportunity to add their thoughts to each topic in a smaller setting. The

    facilitators then work hard (and fast) to summarize all the points and report out to the

    Workshop attendees - all in real time.

    3. The “Plenshop” Format

    Sometimes, a presentation of background material before discussion begins is a good thing.

    The Plenshop Format allows for that. In these sessions, one or more short presentations

    will be followed by intensive discussion. It’s not a plenary session, it’s not a workshop -

    it’s a Plenshop!

    Additionally, we will have a Workshop in the Standard Format which will have the topic chosen

    by WCBP 2016 attendees while at the meeting! We realized that sometimes we may miss an

    important breaking topic because of how far in advance we work to ensure a high-quality

    conference. The Hot Topic Workshop allows attendees to suggest and vote for a topic of their

    choice and we will provide skilled facilitators, whatever the topic! The vote for a “hot topic” will

    be through the Mobile APP. Please make sure to download the APP so that you can cast your

    vote!

    Discussion at Workshops, Round Table Sessions, and during formal and informal networking

    times is critical to the success of our meeting, with the introduction of the new Workshop Formats,

    we hope that you will get even more from WCBP 2016.

  • Workshop Descriptions

    Workshop Session 1

    Tuesday, January 26, 2016

    17:00 – 18:15

    Regulatory and Compendial - Combination Products

    Rakhi Dalal, CDER, FDA, Donna French, Genentech, a Member of the Roche Group,

    Steve Hertz, CDER, FDA, Ed Patton, CBER, FDA, Michael Soberg Christensen, Novo Nordisk

    AG, John Weiner, OCP, FDA

    In the Chinese Room – Standard Workshop

    In January 2015, the FDA issued draft guidance on CGMP requirements for combination products

    in support of the regulation (21 CRF Part 4). These regulations are intended to promote the public

    health by clarifying which CGMP requirements apply when drugs, devices, and biological

    products are combined to create combination products. Combination products are those that consist

    of two or more different types of products, such as a drug/device combo like a pre-filled syringe.

    There many different many different types of combination products and this session can address

    the application of 21 CFR Part 4 to different type of products. This is a challenge for manufacturers

    of combination products as industry has to the responsibility to comply with these regulations and

    enforcement of 21 CRF Part 4 compliance is expected. Clarity of expectations regarding CGMP

    requirements for combination products are crucial for establishing a robust and compliant quality

    system and ensuring a consistent implementation of 21 CFR Part 4.

    This session will address some of the important questions and issues for combination products in

    relation to quality system, experiences/expectations in relation to inspection of combination

    products as well as experiences from the review process. The following specific question will be

    addressed:

    a. How to establish an appropriate quality system for manufacturing of combination products?

    b. How will the interaction between FDA review and inspection teams change future inspections of combination products?

    c. How should design controls and purchasing controls be implemented for various types of combination product technologies (e.g., drugs co-packed with class 1 or class 2 devices)

    d. How do companies meet the requirements for marketed products that were not developed under design controls?

    NOTES:

  • Pay Now or Pay Later: Prioritizing CMC Development Activities in Early Development

    Christopher Downey, CDER, FDA, Catherine Eakin, Seattle Genetics, Michelle Frazier, AbbVie,

    Inc., Sara Gagneten, CBER, FDA,

    In the District Room – Standard Workshop

    With the advent of programs that can rapidly accelerate the normal product development lifecycle,

    such as Breakthrough Therapy Designation, Industry is re-thinking product development strategies

    to achieve shortened timelines. This session will consider the CMC activities that can expedite

    both early and late stage development. Strategies discussed will include the use of platform

    processes and methods, frontloading of characterization studies early in development, and new

    approaches such as the use of pooled clones to enable more rapid entry into first in human

    studies. The risks and benefits of these strategies will be evaluated in the context of technical

    challenges and overall impact to development timelines.

    NOTES:

  • Regulators’ Perspectives on Trends in the Regulation of Biopharmaceutical Products in

    Europe and Asia

    Session Chairs: Kathleen Francissen, Genentech, a Member of the Roche Group and Anthony

    Ridgeway, Health Canada

    Regulators: Peter Richardson, European Medical Association (EMA) and Daisaku Sato, PMDA

    and Anis Talib, Ministry of Health Malaysia (MHM)

    In the Grand Ballroom – Plenshop

    In this session, regulators that represent EMA, PMDA, and MHM will present the highest priorities

    of their agencies and discuss current trends in accelerated development strategies, (such as

    Adaptive Pathways and Sakigake), biosimilars development, and regulatory convergence and

    harmonization (particularly the involvement of their agency).

    Among activities that support regulatory system strengthening, the Asia Pacific Economic

    Cooperation (APEC) Regulatory Harmonization Steering Committee (RHSC) has a mandate to

    work towards achieving regulatory convergence by 2020 among the 21 APEC economies. Japan

    was identified as the new chair of RHSC, and will co-chair RHSC meetings with US FDA. In

    terms of training and capacity building, APEC Training Centers of Excellence (CoE) for

    Regulatory Science, which are partnerships of academia, regulators, and industry (at various stages

    of implementation) identified the following priority work areas: (1) Biotherapeutics; (2) Multi-

    regional clinical trials (MRCT), including Good Clinical Practices (GCP); (3) Pharmacovigilance;

    (4) Good review practices (GRP), including good submission practices; and (5) supply chain

    integrity. The potential opportunities and challenges of achieving regulatory convergence in the

    APEC region and in other regions will be discussed.

    NOTES:

  • Perspectives on Trends in the Regulation of Biopharmaceutical Products in Europe and

    Asia (Japan)

    Daisaku Sato, Pharmaceuticals and Medical Devices Agency (PMDA), Tokyo, Japan

    The Japanese regulation has improved its review performance over the last decade. Bio-

    pharmaceuticals were also the major parts of this expedited program.

    PMDA continues further shortening its review period by 2018. MHLW and PMDA started the

    “Sakigake” (forerunner) review assignment system on a pilot basis in May 2015. It is so called

    break through therapy designation for novel products that are willing to be launched in the

    Japanese market first. More than 50 products/indications were subject to the screening process

    and finally 6 products/indications were assigned to be under Sakigake in October 2015.

    The new legislations for regenerative medicine (cellular and tissue-based products) came into

    effect on 25 November 2014. It introduced early access scheme (conditional and time-limited

    approval) for regenerative medical product to expedite patient access to promising therapies. The

    two products were newly approved under the regenerative medical product category of new

    legislation in September 2015.

    The biosimilar regulations have experienced growing number of consultations during clinical

    trials and two new product approvals in 2015, which are linked with global product

    developments. It may arise the global challenges of selection of comparator reference products

    and statistical evaluation for multi-regional clinical and non-clinical studies. We will present the

    current challenges of biologics CMC review, such as responses to ICH-Q12 guideline to

    accommodate post-approval change in terms of the international harmonization. We will step

    further global collaboration among regulators and biotech industries.

    In Japan, MHLW and PMDA put forward the international cooperation with regulatory

    authorities and set up the international strategic plan in June 2015. It will enable PMDA to

    contribute to the world public health, and to maximize the common benefit through regulatory

    innovation. PMDA has been actively participating in the international harmonization and

    convergence fora including APEC, and promotes bilateral supports to Asian regulators for

    capacity building.

    NOTES:

  • EU Developments in Early Access Programs and International Cooperation

    Peter Richardson, European Medicines Agency (EMA), London, United Kingdom

    In recent years, the EMA and the European Network have been focused on ways to maximise the

    benefit of existing regulatory pathways to facilitate early access to medicines. The EMA has

    offered Adaptive Pathways as an opportunity for industry to benefit from increased regulatory

    interaction by way of a pilot program for medicines with high unmet need. In addition, these

    products should be developed by an iterative process (initially: restricted population or evolving

    surrogate endpoints) and utilising real world data.

    In 2015, the EMA has announced a further t