Presenter to insert their organization’s logo and information here Challenges when implementing guidelines for biosimilars PANDRH, 2013, Ottawa, Ontario.

Slides:



Advertisements
Similar presentations
6th European Patients’ Rights Day The EMA Geriatric Medicines Strategy and the empowered aging patient Francesca Cerreta EMA (European Medicines Agency)
Advertisements

Management of Drug Formulary Dimitry Gotlinsky Western University Managed Care Clerkship ProPharma Pharmaceutical Consultants, Inc. 06/16/06.
Medication Management
AXINN, VELTROP & HARKRIDER LLP © 2007 | AXINN, VELTROP & HARKRIDER LLP Click To Modify Title Name Goes Here FDA Hearings on the BPCI Act.
Session 6 : Need for good governance Importance of veterinary legislation and its appropriate implementation Jean-Luc ANGOT, CVO, France 14 March 2013.
Study Objectives and Questions for Observational Comparative Effectiveness Research Prepared for: Agency for Healthcare Research and Quality (AHRQ)
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
Strengthening the Medical Device Clinical Trial Enterprise
Many Important Issues Covered Current status of ICH E5 and implementation in individual Asian countries Implementation at a regional level (EU) and practical.
UNITED SPINAL ASSOCIATION AUGUST, 2014 Biologics & Biosimilars: An Overview 1.
Physician Perspectives on Subsequent Entry Biologics (SEBs) Michael S. Reilly, Esq. Executive Director, Alliance for Safe Biologic Medicines March 31,
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
The ICH E5 Question and Answer Document Status and Content Robert T. O’Neill, Ph.D. Director, Office of Biostatistics, CDER, FDA Presented at the 4th Kitasato-Harvard.
Christine Simmon Senior Vice President, Policy & Strategic Alliances Generic Pharmaceutical Association November 6, 2014 Biologics Naming.
Career Opportunities for PharmDs in the Pharmaceutical Industry: Research & Development.
Clinical Trials of Traditional Herbal Medicines In India Y.K.Gupta Professor & Head, Department of Pharmacology, All India Institute of Medical Sciences,
Biopharmaceutical Regulatory Requirements 40. Marketing Authorization for New Chemical Entities Health Canada’s (HC) Therapeutic Products Directorate.
Executive summary prepared by some members of the ICH Q9 EWG for example only; not an official policy/guidance July 2006, slide 1 ICH Q9 QUALITY RISK MANAGEMENT.
IPhVWP Polish Presidency, Warsaw October 6 th 2011 Almath Spooner Irish Medicines Board Monitoring the outcome of risk minimisation activities.
Environmental Impact Assessment (EIA): Overview
Subsequent Entry Biologics (SEBs) – Canada Presentation to AIPLA Biotechnology Committee January 25, 2012 Daphne C. Lainson
Data protection and extension of patent rights TRIPS requirements & TRIPS-plus provisions Carlos Correa.
Regulatory Authority Governing Clinical Trials Anthony J. Minisi, MD Director, Cardiology Fellowship Program.
Stakeholders In Clinical Research Government and Regulatory Bodies Professor Phil Warner.
Summary About me & AMRC About the EU Clinical Trials Regulation – what’s new Implementation timeline What does this mean for UK ethicists?
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication A substance used in the diagnosis, treatment,
REGULATORY CHALLENGES FOR NANOMATERIALS IN PUBLIC HEALTH Driving Faster Than Our Nano-Headlights AAAS Annual Meeting February 13, 2009 Norris E. Alderson,
Clinical Trial Review and Approval: New Regulations and their implications Siddika Mithani, Ph.D Clinical Trials & Special Access Programme Therapeutic.
Reproductive Health Drugs, Pregnancy Labeling Subcommittee Meeting March 28-29, 2000 Holli A. Hamilton, M.D., M.P.H. Pregnancy Labeling Team Office of.
Drug Submissions: Review Process Agnes V. Klein, MD Biologics and Genetic Therapies Directorate February, 2003 www/hc-sc.gc.ca/hpb-dgps/therapeut.
CHEE DRUG PRODUCT DEVELOPMENT u Drug ä agent intended for use in the diagnosis, mitigation, treatment, cure, or prevention of disease in man or animals.
Investigational Drugs in the hospital. + What is Investigational Drug? Investigational or experimental drugs are new drugs that have not yet been approved.
Authorisation of medicinal products: selected challenges Rocío Salvador Roldán Pharmaceuticals Unit/DG SANCO This presentation only reflects the views.
1 Department of Medicines Policy and Standards, Health Technology and Pharmaceuticals WHO’s Role in Assuring the Quality Safety and Efficacy of Medicines:
PhAMA Position on Biosimilar Medicines Ms. Leah Goodman.
Marcel H.N. Hoefnagel 2 November 2007 BIOSIMILARS are not Generics But similar.
Issues in Generic Substitution: Safety/Efficacy, Cost Savings and Supply Robert J. Herman, MD, FRCPC Professor, Department of Medicine University of Calgary.
WHO Workshop on Prequalification of Medicines Programme, Abu Dhabi, October, 2010 Regulatory principles reflected in practice of WHO PQP Milan Smid,
Biologics and Genetic Therapies Directorate Health Products and Food Branch HEALTH CANADA Helping Canadians maintain and improve their health. Agnes Klein.
The New Drug Development Process (www. fda. gov/cder/handbook/develop
History of Pediatric Labeling
Inclusion of Women in Clinical Trials & Drug Development Regulatory Perspective Ameeta Parekh, Ph.D. R&D Director Office of Women’s Health Food & Drug.
International Atomic Energy Agency Roles and responsibilities for development of disposal facilities Phil Metcalf Workshop on Strategy and Methodologies.
Is the conscientious explicit and judicious use of current best evidence in making decision about the care of the individual patient (Dr. David Sackett)
Risk Sharing Schemes Dr Rafiq Hasan Director of Market Access
European Patients’ Academy on Therapeutic Innovation Ethical and practical challenges of organising clinical trials in small populations.
1 PRIORITY MEDICINES FOR EUROPE AND THE WORLD Barriers to Pharmaceutical Innovation Richard Laing EDM/PAR WHO.
Overcoming challenges in pediatric oncology product development: Regulatory oversight of multi-national clinical studies Ursula Kern, Advisory Committees.
Abstract A step-wise or ‘tiered’ approach has been used as a rational procedure to conduct environmental risk assessments in many disciplines. The Technical.
Compassionate use programs and the European regulatory system Filip Josephson M.D., Ph.D. Clinical Assessor.
Final Canadian Guidelines for “Biosimilars” Subsequent Entry Biologics (SEBs) DIA RA SIAC RD/RI Working Groups 11-May-2010 Stephen Sherman.
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication recognized or defined by the U.S. Food, Drug,
Storage, Labeling, Controlled Medications Guidance Training CFR § (b)(2)(3)(d)(e) F431.
Continuous Improvement & Real World Evidence: A Public Payer’s Perspective Suzanne McGurn, Assistant Deputy Minister and Executive Officer Ontario Public.
Drug Development Process Stages involved in Regulating Drugs
RAC Regulatory Affairs Certification
Periodic Safety Update Reports (PSUR)
FDA’s IDE Decisions and Communications
Karen Proud, President Consumer Health Products Canada
REGULATORY PROBLEMS IN CARING OUT PRE- AND POST- AUTHORISATION CLINICAL TRIALS Dr Penka Decheva GCP Inspector, BDA.
Pharmacovigilance in clinical trials
Medical Device Regulatory Essentials: An FDA Division of Cardiovascular Devices Perspective Bram Zuckerman, MD, FACC Director, FDA Division of Cardiovascular.
. Regulatory Approach to Subsequent Entry Biologics in Canada
WHO’s Role in Assuring the Quality Safety and Efficacy of Drugs:
ADVAC ALUMNI MEETING DURING SAGE
Prescription-only vs. over-the-counter medicines
Pharmaceuticals Industry
Regulatory Perspective of the Use of EHRs in RCTs
Assessing Similarity to Support Pediatric Extrapolation
Presentation transcript:

Presenter to insert their organization’s logo and information here Challenges when implementing guidelines for biosimilars PANDRH, 2013, Ottawa, Ontario Agnes V. Klein, MD DPH Director, Centre for Evaluation of Radiopharmaceuticals and Biotherapeutics,

2 Overview General considerations:  Regulation of the life-cycle of biological drugs in Canada  Regulatory context of biosimilars  What types of challenges has Canada encountered and continues to see in the implementation of biosimilar guidelines? Challenges and post-market issues of “older biosimilars”  Are there several “types of biosimilars” ?  How can regulators have confidence in the quality, safety and efficacy of “older biosimilars”?  How can the gaps be filled?  Conclusions

3 Regulating the life-cycle of biological drugs  Drugs are “living entities” and have a life-cycle that goes from discovery, through development, use for treatment of disease and, finally, in some instances either removal from market due to safety considerations or due to or lack of use  Increasingly, it has become clear that this life-cycle must be managed to develop and maintain their benefits and risks at usable levels and in usable form: benefits outweigh risks  Life-cycle management of drug products is similar for originators as well as for generics and biosimilars  The newest PBRER ICH Guideline (ICH E2C (R2) has been adopted by Canada to manage all drugs, including biologicals, following their market authorisa tion

REGULATORY CONTEXT OF BIOSIMILARS  In Canada biosimilars are called Subsequent Entry Biologics (SEBs)  They are subject to the same regulations and processes as originator biologic drugs  A framework guideline provides appropriate interpretation and the context within which biosimilars are regulated  The Federal regulator in Canada authorises drugs for market  Regulated as drugs under new drug regulations  Intellectual property follows data protection provisions similar to the provisions for generics  Due to their complex nature there are special considerations for extrapolation of indications  There is no declaration of substitutability: this is a provincial matter and an issue related to the practice of medicine 4

ARE THERE SEVERAL “TYPES OF BIOSIMILARS” ?  Under Canadian Regulations there is only one type of biosimilar or SEB drug  In some jurisdictions the “biosimilar” nomenclature applies or applied to a number of biologicals considered essentially identical to the molecule of the originator  However, biologicals can never be reproduced as identical molecules and the concept of identity does not apply  At minimum, there are unknown risks between various lots and active ingredients considered to be the same  Therefore, these molecules may be considered only as new biologicals that were marketed without a complete safety, efficacy and quality package. For these the interplay of the three elements were never researched: no data of value exists in respect of these products (literature and theoretical information only) 5

CHALLENGES IN IMPLEMENTING BIOSIMILAR GUIDELINES  Manufacturing and pharmaceutical challenges:  The source of the API could give rise to impurities that differ from the originator  The formulation, while equivalent may have a bearing on the PK/PD properties and, therefore, the dosage of the final product  Comparability between the RBP and the biosimilar may not be the same from lot to lot: drifts over time are possible  The RBP chosen is not a Canadian product and differs in dosage form, and/or concentration, and/or mode of administration, from those of the biosimilar.  There are no bridging rationales in respect of the Canadian product, or a foreign-marketed product; when necessary, studies are missing as sponsors did not consult in advance on requirements Two biological products meet the standard of being truly similar only at the time of their initial market authorisation 6

CHALLENGES IN IMPLEMENTING BIOSIMILAR GUIDELINES  Safety challenges:  Is the potential toxicity or safety of the product similar to that of the RBP?  The immunogenicity of the two products, the RBP and the biosimilar will likely differ (for ex: EPO)  Switching from the RBP to a biosimilar can elicit immune phenomena that were not foreseen  We worry mostly about the systemic, generalised immune reactions/secondary diseases  Safety-related matters may also have unexpected consequences, clinically 7

CHALLENGES IN IMPLEMENTING BIOSIMILAR GUIDELINES Naming and Traceability for Biosimilars 8 A biosimilar should have its own brand name (trade name) as for all new medicinal products International Non-proprietary Name (INN) is within responsibility of WHO. HC currently follows WHO policy, but further discussion is needed on what triggers unique INN Brand name, INN, DIN, lot/batch number and manufacturer of biological products should be included in adverse reaction reporting

CHALLENGES IN IMPLEMENTING BIOSIMILAR GUIDELINES  Efficacy challenges:  Difficulties in decision-making when design is not an equivalence trial and margins of equivalence have not been predefined  Outcomes of non-inferiority trials not always as expected:  Lead to difficulties around efficacy  Acceptable for safety and immunogenicity  Considerations around extrapolation of indications  Study design not suitable and/or rationale not provided  Considerations missing often: pathophysiology of disease/s, patient population enrolled not suitable, PK/PD differences not accounted for, questions of dosage, route of administration, dose ranges, mono or combination therapy  Age, sex, race, genomics, not considered  Duration of study too short, 9

CHALLENGES IN IMPLEMENTING OF BIOSIMILAR GUIDELINES  There may be other challenges as yet not encountered, including lack of data  We are still in our “learning phase” but are taking a pragmatic and operational approach to resolve these until we have learned a sufficient amount to update our guidelines  We are, however, very careful to be consistent with all decisions  These decisions must apply to both biosimilars and to new manufacturing methods for originator biologicals 10

“OLDER BIOSIMILARS”: CONFIDENCE IN THEIR QUALITY, SAFETY AND EFFICACY  S ome countries/regulators have biosimilar products that were never compared to a fully developed originator  Indications and uses have been “copied” from class indications in “benchmarking” countries  Very little is known about these products  Their scientific and medical status is uncertain, but their regulatory status is even more uncertain  Consequences are gaps on efficacy and more importantly, on risks both real and perceived. The nature of theses risks is unclear and/or unknown 11

HOW CAN THE GAPS BE BRIDGED?  Risks can be evaluated and categorized  The type of categorization will depend on the regulations existing in each country  Guidelines may be available already. If not, guidelines could be drafted to fit with general scientific and risk- assessment principles that will allow the risk of each product to be evaluated  Stakeholders should play a role in all of these activities: consultation is important  Guidelines should contain the minimal requirements for updating of information, especially on the safety side. ICH E2c(R2) can serve as a basis 12

HOW CAN THE GAPS BE BRIDGED?  ICH E2C (R2) deals with PBRERs: Periodic benefit-risk assessments for products on the market  It is important to provide time-lines (sufficient time) for updating information and, if necessary, removal of products from market within each country’s regulatory authority  Overall knowledge of the class of product can be also used to help define risks and time lines  Use of international information such as warning letters issues by other regulators, etc. can be used to help in these risk evaluations and subsequent course of action  Standard Operating Procedures and templates for recording information can be very helpful and ensure consistency 13

HOW CAN THE GAPS BE BRIDGED?  It is also useful to have annual reports on the quality parameters for each product  PBRERs will provide information and an understanding of the ongoing benefits and risks of a product. It is important to keep in mind that perspectives and points of view of all concerned groups may not be the same  Industry and Partners (development)  Regulator (independent assessment/decision)  Patients, Payers, Stakeholders (real-world experience)  A learning period is important in all new activities: biosimilars are no exception 14

HOW CAN THE GAPS BE BRIDGED? One practical example:  A biological was seen to have several risks with uncertainties: manufacturing, safety and labelling  A risk assessment was conducted  Consultations were undertaken (web-based)  The risk level was not defined but it became clear that information gaps existed that needed to be bridged  Sponsors were given up to 2 years to bring the information behind the efficacy, safety and quality of their products up to date  The process is now ongoing and we are receiving complete submissions 15

CONCLUSIONS  Life-cycle management of drug products is similar for originators as well as for biosimilars  Biosimilars are subject to the same regulations and processes as originator biologic drugs  Biologics can never be reproduced as identical molecules and the concept of identity does not apply  Two biologic products meet the standard of being truly similar only at the time of their initial market authorization  When making changes, appropriate risk assessment, management and communications principles should be considered and followed  International guidelines are helpful:  PBRERs will provide information and an understanding of the ongoing benefits and risks of a product. It is important to keep in mind that perspectives and points of view of all concerned groups may not be the same  Stakeholders should be consulted for their perspectives  Timelines for implementation are very desirable 16

Thank You ? 17