Comparability for Biotech Products

Download Report

Transcript Comparability for Biotech Products

Comparability for Biotech Products: An industry perspective on past & future aspects

Mary B. Sliwkowski, Ph.D.

VP, Regulatory CMC & Info Systems Company logo here

Disclaimer

The views and opinions expressed in the following PowerPoint slides are those of the individual presenter and should not be attributed to Drug Information Association, Inc. (“DIA”), its directors, officers, employees, volunteers, members, chapters, councils, Special Interest Area Communities or affiliates, or any organization with which the presenter is employed or affiliated. These PowerPoint slides are the intellectual property of the individual presenter and are protected under the copyright laws of the United States of America and other countries. Used by permission. All rights reserved. Drug Information Association, DIA and DIA logo are registered trademarks or trademarks of Drug Information Association Inc. All other trademarks are the property of their respective owners.

Agenda

• Comparability: what, why, when, how • Experience so far • Challenges & lessons learned • The future

Why do we need ‘comparability’?

• Small molecule drugs – Fully defined by physico-chemical methods • Biotech products – Complex mixtures of 100s - 1000s of forms – Post-translational modifications – Higher order structure – Defined by characterization & process • Physico-chemical • Biologic (in vitro) • Animal (pharmacokinetics, toxicology, immunogenicity) • Human (safety, efficacy) • Process qualification & validation – Well-characterized but not fully defined

Comparability: What is it?

• “Comparability is the demonstration of a high degree of similarity between products produced by different manufacturing processes, equipment and/or sites, such that no adverse impact on quality, safety or efficacy occurs” • Comparability is NOT identity

Comparability: Global Guidance

• Comparability is a well established global regulatory mechanism based on ICH Q5E

“Comparability of Biotechnological / Biological Products Subject to Changes in Their Manufacturing Process”

• Per ICH Q 5E: – “The goal of a comparability exercise is to ascertain that pre and post-change product is comparable in terms of quality, safety and efficacy ” – “The demonstration of comparability does not necessarily mean that the quality attributes of the pre-change and post-change are identical, but that they are highly similar …”

Comparability: History of U.S. Biotech Regulation

1980

• •

First Biotech Drugs Insulin Growth hormone *PTC for Biotech 1986 ICH PDUFA I REGO PDUFA II FDAMA PDUFA III CBER/CDER Integration PDUFA IV First Biotech Biologics 1990 CBER/CDER Inter-center Agreement

• • • • •

‘92 ‘94 ‘97 ‘98 2000 ‘02 ‘03

• •

Comparability Guidance Comparability Protocols First WCBP Symposium Specified Products BLA replaced PLA & ELA Eliminated FDA Lot Release Created Team Biologics ‘04 ‘05 ‘06 ‘07

• • • •

Quality by Design Design Space Process Analytical Technology (PAT) Expanded Change Protocols

Regulatory Mechanisms

• Post-Approval Submission (PAS) – Conduct qualification of change and submit data for approval prior to implementation • Comparability Protocol – Pre-specify testing, validation studies and acceptance criteria for change to be made – Pre-change approval may allow reduced reporting category when implement change – Initially only for specific, pre-defined change to single product – Expanding to broader applications - multi-change, multi-product • Expanded Change Protocol (TBD) – Linked to Quality by Design approach (ICH Q8) – Provide evidence of sufficient product knowledge (Critical Quality Attributes - CQAs) and process understanding (Critical Process Parameters - CPPs) to define a Design Space in which can operate more freely • EU Variation regulations being revised

Comparability: Throughout the Product Lifecycle

Clinical Development

• some product variability (lot-to-lot, inter-campaign) is desirable • process changes expected • collecting clinical and non-clinical data

Phase III – Registration

• process, control system, formulation locked • collecting pivotal clinical data • qualification, validation lot production • process changes will require BE and/or efficacy data

Post-Approval

• apply comparability guidance (ICH Q5E) • match historical data sets

What triggers comparability efforts?

• Manufacturing facility changes – Site, scale, equipment • Process changes – Cell culture, fermentation • Cell line changes – Recovery – Formulation – Delivery mechanism or system • Supplier changes – Raw materials – Primary components • Regulatory expectation changes

Hierarchy of Comparability Testing

Category

A - Basic Package B - Biological Characterization C - Animal PK/PD D - Human Bioequivalence E - Human Clinical Trials

Testing

Determined on case by case basis • CoA • Extended characterization of variants & impurities • Accelerated degradation • In vitro functional bioassays & binding studies • Real time & accelerated stability • Rodent PK • Primate PK/PD • Direct comparison of pre- and post-change • Adequately powered • To confirm efficacy, safety, and/or immunogenicity

Chronology of Key Biotech Product Approvals

1982 - 2007

Follistim Forteo

Humulin R

(1982) Humatrope Glucagon Thyrogen Mylotarg Ovidrel Increlex Nutropin Protropin Ceredase Cerezyme Natrecor 85 86 87

Intron A

88 Activase 89

Epogen /Procrit

CDER (N=14) CBER (N=48) 90 91 92 93 94 95 96 97 98 99 00 01 02 03 04 05 06 07 Proleukin

Neupogen

Leukine Actimmune Pulmozyme Betaseron ReoPro NovoSeven Ontak

Avonex

Retavase Benefix Infergen Neumega

Rituxan

Zenapax Regranex Simulect Synagis Remicade

Herceptin Enbrel

Refacto Campath Kineret Xigris Aranesp > $1 Billion Annual International Sales Fabrazyme Amevive Xolair Bexxar Raptiva Zevalin Elitek

Humira

Rebif Lucentis Myozyme Elaprase Vectibix Abatacept Galsulfase

Avastin

Erbitux Tysabri

What is the GNE experience

• 23 years of commercial manufacture • 14 products with 8 process version changes • Drug substance transfers/sites completed – Within GNE network: 14 across 4 sites – Out to partners/CMOs: 8 across 6 sites – In to GNE: 1 at 1 site • Drug Product transfers/sites completed – Within GNE network: 10 across 3 sites – Out to partners/CMOs: 11 across 8 sites • Submitted 13 PAS & 12 CP • Many transfers planned as rebalance mfg network • GNE internal documents: Quality & Regulatory Standards

What have we learned

• Don’t make process changes between Phase III and approval • Platforms have limitations – Each Mab has unique challenges • Trastuzumab: Met oxidation, Asn deamidation • Omalizumab: unpaired Cys, Asp isomerization • Bevacizumab: dissociable aggregates • Ranizumab: Trp oxidation • Sequence variants can/do occur • Minor carbohydrate variation is sometimes important • Relevance of differences is situational • Maintain frequent dialog with Agency • Successful comparability efforts require due diligence • Our ultimate responsibility is to our patients

Challenges

• Evolution of analytical methods – Increased sensitivity – New forms • Higher order structure • Linkage to safety & efficacy – When is a difference significant?

– Mechanism of action – Patient population – Route of administration • Comparability is still case by case • Global regulatory differences – Change regulations mechanisms – Stability requirements – Timing of transition

Acknowledgements

Wassim Nashabeh Kathy Francissen Reed Harris Amita Joshi Ron Taticek Ray Arnold Loel McPhee