Basis for setting acceptance criteria

Similar documents
Clinical qualification of specifications - a Regulator s view

Report on the expert workshop on setting specifications for biotech products

Setting Specifications for Biotech Products

Recent experience in scientific advice and marketing authorisations

European Medicines Agency Evaluation of Medicines for Human Use

Control strategy and validation. Emanuela Lacana PhD Office of Biotechnology Products CDER/FDA

Biosimilar Monoclonal Antibodies: Registration Requirements. Henry M. J. Leng

Experience of Post approval change management protocol in EU. Mats Welin Senior expert Medical Products Agency Uppsala, Sweden

Regulatory perspective on setting clinically relevant specifications. Joslyn Brunelle, PhD Team Leader Office of Biotechnology Products

Using prior knowledge in applications. Mats Welin Senior Expert Medicals Products Agency, Uppsala, Sweden

Regulatory Perspective on Analytical Method Validation During Product Development

Preparing the CMC section of IMPD for biological/biotechnology derived substances

Ph. Eur. monographs and biosimilars

Regulatory Challenges for the Licensure of Future Vaccines

Documentation requirements for an initial consultation

Guideline on Similar Biological Medicinal Products

GUIDE TO GOOD MANUFACTURING PRACTICE FOR MEDICINAL PRODUCTS

Guideline on the Requirements for Quality Documentation Concerning Biological Investigational Medicinal Products in Clinical Trials

Reflection paper on the requirements for selection and justification of starting materials for the manufacture of chemical active substances

Examples of regulatory expectations for analytical characterization and testing

Guideline on the requirements for quality documentation concerning biological investigational medicinal products in clinical trials

Writing a Convincing Dossier on Impurities and Method Validation. Dr. Hans Ulrich Gally Swissmedic Swiss Agency for Therapeutic Products

Guideline on the requirements for quality documentation concerning biological investigational medicinal products in clinical trials

Phase Appropriate GMPs for IMPs. Presented by: Karen S. Ginsbury For: IFF, October 31, Nov 02, 2017

Justification of Specifications (JOS): What Product and Process Development was all About. Christopher A Bravery

Consultation by a notified body on an ancillary medicinal substance integrated in a medical device

Challenges with Establishing a Control Strategy for Biosimilars

Good Manufacturing Practices Purpose and Principles of GMP. Tony Gould

Risk Assessments for Host Cell Protein Control Strategies: CDER Experiences

Guideline on the requirements for quality documentation concerning biological investigational medicinal products in clinical trials

WHO PQ dossier Module I. DCVMN Common Technical Document (CTD) Workshop Brazil, 18 to 20 June 2018 Dr. Nora Dellepiane

London, 11 October 2006 Doc. Ref. EMEA/CHMP/BWP/271475/2006 COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

What is comparability? An updated perspective. Martijn van der Plas MEB

1225 Eye Street NW, Ste. 400 Washington, DC 20005

Harmonized Quality Requirements for Biotech Investigational Medicinal Products - A Regulator's View

2017 AAM CMC Workshop

Process Performance Monitoring & needed engagement tools

Phase-Appropriate Analytical Method Validation: A Regulator s Perspective

Recent Trends in the Evaluation of Analytical Biosimilarity

Regulatory Consideration for Biotechnology Products: Clonality of the Production Cell Bank

The importance of interchangeability in the procurement of medications: Biosimilar case

Update on the new immunogenicity guideline in the EU

Regulatory Issues and Drug Product Approval for Biopharmaceuticals

ANALYTICAL VALIDATION CHALLENGES DURING THE RAPID DEVELOPMENT OF KEYTRUDA

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS NOTE FOR GUIDANCE: MANUFACTURE OF THE FINISHED DOSAGE FORM

Regulatory expectations on impurities in drug substances - Pavia, October 2, Luisa Torchio Euticals SpA

How the European Pharmacopoeia Provides the Framework to Implement QbD Principles

ICH Q11 Development & manufacture of drug substances

Ames Data Submissions and Other Qualification Data for Impurities in Drug Substances

EudraLex. Part I1 Chapter 6: Quality Control

VALIDATION OF ANALYTICAL PROCEDURES: METHODOLOGY *)

Regulatory Assessment

EUROPEAN COMMISSION HEALTH AND CONSUMERS DIRECTORATE-GENERAL. EudraLex. The Rules Governing Medicinal Products in the European Union

Quality aspects Overview. QP declaration

Comparability to establish Biosimilarity

ICH Topic M 4 Q Location issues for Common Technical Document for the Registration of Pharmaceuticals for Human Use Quality Questions and Answers

Non-clinical documentation Overview of Requirements

COMMERCIAL PRODUCT STABILITY

Chemical Aspects of Stability Evaluation

Validation of Analytical Methods used for the Characterization, Physicochemical and Functional Analysis and of Biopharmaceuticals.

Wrap Up Summary CMC Strategy Forum. Bridging Analytical Methods Jan 27, 2014

CASE STUDY: THE USE OF PRIOR KNOWLEDGE IN ESTABLISHMENT OF AN INTEGRATED CONTROL STRATEGY AND CLINICALLY RELEVANT SPECIFICATIONS

Guidance for Industry Q3B(R2) Impurities in New Drug Products

COMMON DEFICIENCIES IN FINISHED PHARMACEUTICAL PRODUCT (FPP) DOSSIERS

Questions and answers on the 'Guideline on the limits of genotoxic impurities'

KFDA Regulatory Framework on Biopharmaceuticals - Focus on Biosimilar

Biosimilar mabs Clinical issues Regulatory perspective

EUROPEAN COMMISSION ENTERPRISE AND INDUSTRY DIRECTORATE-GENERAL. EudraLex The Rules Governing Medicinal Products in the European Union

FDA s Implementation of the Legal and Regulatory Framework for Biosimilars

DRAFT OECD GLP ADVISORY DOCUMENT NO. 19 ON THE MANAGEMENT, CHARACTERISATION AND USE OF TEST ITEMS PREAMBLE

Being Clinically Relevant While Setting Specifications

Specifications. February 19, 2015, TOKYO, JAPAN. Takao Hayakawa, Ph.D. Director, Pharmaceutical Research and Technology Institute, Kindai University

UNICEF s Quality Assurance System for Procurement of Finished Pharmaceutical Products

QUALITY OF PROLONGED RELEASE ORAL SOLID DOSAGE FORMS

Quality Issues for Clinical Trial Materials: The Chemistry, Manufacturing and Controls (CMC) Review

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

Notice Our file number:

PROCESS VALIDATION ROCHAPON WACHAROTAYANKUN, PH.D.

Development of non-substantially manipulated cell-based ATMPs 1 : flexibility introduced via the application of the risk-based approach

European Regulatory Experiences and Expectations of HCP Analysis and Control

Pharmacist Rana Musa Al-ali (Malkawi) MSc (Pharmaceutical Quality Assurance) Registration Department/JFDA

A holistic regulatory approach to accelerated CMC development

Quality Assurance in Pharmaceutical and Biotech Industries as Per Regulatory Guidelines

Considerations in Setting Specifications

ICH 교육가이드라인 [ 안전성 & 품질 ]

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

CMC Strategy Forum Europe May Killarney, Ireland.

Roadmap of stability studies for Biosimilarproduct development. Head Biotech Stelis Biopharma

Guidelines on Similar Biologic: Regulatory Requirements for Marketing Authorization in India

INGREDIENTS, FOOD SUPPLEMENTS AND NUTRITIONAL

Guidance for Industry Process Validation: General Principles and Practices. F. Hoffman-La Roche, Ltd.

Mitigating Commercial Risks in Continuous Manufacturing Drug Product

Affigenix Biosolutions Private Ltd, 265/ 1F, KSSIDC Industrial Area Bangalore, Karnataka,India

Adopted by CHMP for release for consultation 15 December Start of public consultation 15 December 2016

ICH guideline Q11 on development and manufacture of drug substances (chemical entities and biotechnological/biological entities)

Data Integrity Why is it important? DADM Conference - 22 August 2017

DJ Cockburn Head of Manufacturing and Quality Compliance European Medicines Agency

GUIDELINE FOR THE STABILITY TESTING

Transcription:

Basis for setting acceptance criteria Mats Welin, Senior expert Medical Products Agency, BWP Disclaimer: The views presented in the presentation are my own and not necessarily the ones of BWP

Fundamentals Q6B Characterization of a biotechnological or biological product by appropriate techniques is necessary to allow relevant specifications to be established. Acceptance criteria should be established and justified based on data obtained from lots used in preclinical and/or clinical studies, data from lots used for demonstration of manufacturing consistency and data from stability studies, and relevant development data.

Purpose of limits/specifications Confirm intended potency Confirm absence of critical contaminants and impurities Confirm that the manufacturing process is under control (also to mitigate the risk of the unknown ) Confirm that released lots are similar to lots tested in clinical trials

Dilemma How can we find acceptance criteria that do not exclude good batches because of normal variation but still is able to pick up outliers that are results of deviations in manufacturing process, starting material etc? Based on characterisation studies appropriate, robust methods are chosen to mirror key aspects of the product in question. As a consequence deviations from the normal in tested attributes may, even if within qualified limits, indicate deviations in attributes not tested for.

Regulatory concerns Clinical qualification are considered the most important aspect when setting the acceptance criteria. Acceptance criteria applied for critical attributes should not be wider than what has been clinically qualified. Acceptance criteria for non-critical attributes can be based on process capability allowing wider limits than what have been used in the clinical trials

Key issue Often only few batches are used in the clinical trials and these may not cover the full span of normal production variability. How to reach acceptable limits to avoid unnecessary rejection and still be able to claim clinical justification?

Considerations What prior knowledge (e.g. platform knowledge) can be used in claiming clinical justification? Not all clinical studies look at relevant parameters (e.g. immunogenicity) to verify if certain product related forms gives rise to antibody formation or not. Dose finding studies can be useful- even if the content of a particular impurity expressed in % may be below the proposed limit, the actual exposure should be accounted for (e.g. results from 2x dose studies may justify a higher % than used in the studies of the chosen dosage)

Considerations cont. Non-clinical data could be useful in justifying acceptance criteria but normally the concern in relation to impurities in biotech products relates to immunogenicity rather than toxicity and the value of non-clinical data are therefore limited. Could be used for other aspects though Drift in e.g. glycosylation or charge pattern where a form already exposed to humans increases or decreases may be less critical than the appearance of a completely new form.

Pitfall Validation should show that commercial process result in a product that falls within clinically qualified limits, not the specifications in force during the clinical trials which may be much wider. Reference to acceptance criteria applied in the clinical trials is not sufficient to justify commercial acceptance criteria.

Use of statistics in claiming qualification. Proposals have been made to apply method variability to results seen in clinical trials- e.g. clinical trial point estimates +/- 3 SD claiming that the true value of the batch used in the studies could equally well have been something else. True, but could also equally well have been something on the other side of the span i.e. the result seen in the trial material could be the true value + 3SD.

Use of statistics in claiming qualification cont. It is not deemed appropriate to add method variability as determined in validation studies to the variation seen in batch results ( i.e. specification = batch results +/- 3SD + method variability) when justifying a specification as this variability is already included in the batch results and therefore will be counted twice

Use of stability results in setting acceptance criteria By default used when changes are seen during storage to set release limits to assure a clinically qualified shelf life limit It is not appropriate to add results from stability studies (t=3,6,9 months etc) of the same batch to the pool of batch analysis results in setting acceptance criteria at release even if no changes are seen during storage as these results are only showing the method variability and not the sum of variability of a particular attribute (content etc.) and its test method. Could however be useful to check variability over time. Age of material used in clinical trials can be used in justification of acceptance criteria.

Ph Eur Ph Eur mandatory if monograph exists. to be considered as minimal requirementsshould be fulfilled but stricter requirements may be required e.g. Ph Eur monograph fulfilment is not sufficient for biosimilarity claim. Consequences for stand alone products?

Key question Is it possible to separate clinical justification and consistency in setting acceptance criteria? To base the regulatory requirements on the clinically qualified levels To handle consistency (process capability) through the company s PQS (e.g. alert/ action levels) where the principles are described in the dossier ( e.g. what are the principles applied in the assignment of the alert/action levels and what kind of action is taken if results are outside these limits) to assure regulators that things are under control. Not to be considered as an overall approval of the PQS as such as this is GMP

Thank you for listening