Examples of regulatory expectations for analytical characterization and testing

Similar documents
Regulatory Perspective on Analytical Method Validation During Product Development

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

Analytical Methods Development and Validation

APPLYING SCIENTIFIC CONSIDERATIONS AND STATISTICAL APPROACHES IN ANALYTICAL SIMILARITY ASSESSMENT

Setting Specifications for Biotech Products

Impurities from degradation of Drug Substances

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

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

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

Strategies for IND Filing Success: Chemistry, Manufacturing and Controls

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

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

Cell Therapy Product Manufacturing Considerations. July 17, 2017 CMC Strategy Forum Mo Heidaran, Ph.D.

A.1 Contents file 4 to 5 A.1 (1)

European Medicines Agency Evaluation of Medicines for Human Use

Validation/Verification of Test Methods An FDA Perspective. Laure H. Kairawicz, Ph.D. Senior Scientist Expert Witness

Notice Our file number:

Scientific and Regulatory Perspectives on Measuring Protein Aggregation of Biotechnology Products

VICH Topic GL2 (Validation: Methodology) GUIDELINE ON VALIDATION OF ANALYTICAL PROCEDURES: METHODOLOGY

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

Replacing Analytical Methods for Release and Stability Testing A Regulatory Perspective

Recent experience in scientific advice and marketing authorisations

VALIDATION OF ANALYTICAL PROCEDURES: METHODOLOGY *)

LAB EXPERTS AT YOUR SIDE Over twenty years of experience

API Stability Protocols and. Chris Byrne Tasmanian Alkaloids

Drug Product Comparability: Using Process and Product Knowledge for Successful Comparability Exercises

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

Early Development Best Practices for Stability- Regulatory Perspective

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

Replacing Analytical Methods for Release and Stability Testing CBER Perspective

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE GUIDELINES INDEX

Stability of Biological Products

BIOEQUIVALENCE TRIAL INFORMATION FORM (Medicines and Allied Substances Act [No. 3] of 2013 Part V Section 39)

Critical Quality Attributes for Biotechnology Products: A Regulatory Perspective

Validation, Verification and Transfer of Analytical Methods (Understanding and implementing guidelines from FDA/EMA, USP and ICH)

Risk Assessments for Host Cell Protein Control Strategies: CDER Experiences

ASMF/DMF Quality Assessment Report (QAR) IGDRP Quality Working Group

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

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

BIOEQUIVALENCE TRIAL INFORMATION

Analytical Procedures and Methods Validation for Drugs and Biologics

Impurity Control in the European Pharmacopoeia

Clinical qualification of specifications - a Regulator s view

Regulatory considerations for Global Haplobank

Guidance for Industry

Regulatory Issues and Drug Product Approval for Biopharmaceuticals

Pharmaceutical Reference Standards: Overview and Role in Global Harmonization

Method Validation Studies How GLP Interacts With Guidance Documents

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

Analysis of Protein Biopharmaceuticals

Comparability Is Not a Nightmare, Just Think Ahead!

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

CMC Strategy Forum, Paris 2008

Regulatory Expectations for Method Transfers: Health Canada's Perspective CMC Strategy Forum Methods on the move January 23, 2017

ICH 교육가이드라인 [ 안전성 & 품질 ]

Regulation of Active Pharmaceutical Ingredients (API)

Perspectives on Method Validation: Importance of Adequate Method Validation

Drug Development - Points to Consider

ICH Q11 Development & manufacture of drug substances

Introduction to CMC Regulatory Affairs

ICH Q2(R1) A Primer. cgmp. GxP PIC/S SOP ISO QA/QC LOD/LOQ. Validation of Analytical Methods API EP FDA OECD GCP

Understanding the Characteristics and Establishing Acceptance Criteria for Analytical Methods Validation

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

The Role of Mass Spectrometry for Developing Biotherapeutics: Regulatory Perspectives

How to Avoid Common Deficiencies in INDs and NDAs. Ramesh Raghavachari, Ph.D. Branch Chief, Branch IX ONDQA/OPS/CDER

On the Q&A to the Guideline for Common Technical Documents

Comparability to establish Biosimilarity

Dr. Earl Dye CMC/GMP Considerations for Expedited Development Programs

European Regulatory Experiences and Expectations of HCP Analysis and Control

Phase Appropriate Method Validation

ICH guideline Q12 on technical and regulatory considerations for pharmaceutical product lifecycle management

QbD Concepts Applied to Qualification and Transfer of Analytical Methods

2017 AAM CMC Workshop

Analytical Procedures and Methods Validation for Drugs and Biologics

Changes to a Potency Bioassay for Biotechnology Products: a Regulatory Perspective Kathleen A. Clouse, Ph.D., Director Division of Monoclonal Antibodi

Medicines Control Authority Of Zimbabwe

Chemical Aspects of Stability Evaluation

Expectations for Analytical Characterisation in the Evaluation of Biosimilarity: A Regulator`s Perspective

Documentation requirements for an initial consultation

IMPURITIES IN NEW DRUG PRODUCTS

á1225ñ VALIDATION OF COMPENDIAL PROCEDURES

ICH Quality Implementation Working Group POINTS TO CONSIDER

Being Clinically Relevant While Setting Specifications

LEGAL REQUIREMENTS FOR STABILITY

Validation & Transfer of Methods for

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

Common Issues in Qualification and Validation of Analytical Procedures

Control Strategy. Implementation of ICH Q8, Q9, Q10

SESSION 11 Stability Data Evaluate, Set Specifications and Prepare Reports

ANTIBODY DRUG CONJUGATES AND BISPECIFIC ANTIBODIES: SCIENTIFIC & REGULATORY CHALLENGES AND OPPORTUNITIES

FDA S GUIDANCE FOR INDUSTRY ANDAS: STABILITY TESTING OF DRUG SUBSTANCES AND PRODUCTS

Kirsten L. Vadheim, Ph.D. BioCompliance Consulting, Inc. Presented at the ASBM Forum, February 25, 2014, Washington DC

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

Validation of Impurity Methods, Part I

参考資料. Joint MHLW/EMA reflection paper on the development of block copolymer micelle medicinal products. Draft

Joint MHLW/EMA reflection paper on the development of block copolymer micelle medicinal products

ICH Q11 Questions & Answers Selection & Justification of Starting Materials. Step 4 August Implementation Working Group

Primary / secondary standards in pharmaceutical QC. Dr. Christian Zeine LGC Standards GmbH. Webinar Series 2013 July 2013

Transcription:

Examples of regulatory expectations for analytical characterization and testing AT Europe 2016, 18 March 2016 Vicki Venizelos Quality RA B.V. Leiden, the Netherlands

Overview What are the regulatory expectations? o Overview of global guidelines o Product life cycle o Analytical qualification/validation requirements What did the Health Authorities expect? o Examples 2

What do regulators expect? ICH Guidelines (EU, US, Japan): o Q1A(R2) o Q2(R1) o Q5C o Q5E o Q6B o M4Q(R1) o. Stability Testing of New Drug Substances and Products Validation of Analytical Procedures: Text and Methodology Stability Testing of Biotechnological/Biological Products Comparability of Biotechnological/Biological Products Subject to Changes in their Manufacturing Process Specifications : Test Procedures and Acceptance Criteria for Biotechnological/Biological Products The Common Technical Document for the Registration of Pharmaceuticals for Human Use: Quality (see References slide) 3

Analytical methods are used for: Product life cycle - I o Preclinical development Support manufacturing development Testing of preclinical batches Stability testing to support first-in-human clinical trials o Clinical development (Phase I/II, III) Support manufacturing development Characterization studies Release testing of clinical batches Stability testing Comparability 4

Product life cycle - II Analytical methods are used for: o Commercialization Validation of manufacturing process Characterization of key intermediates, DS, DP Release testing of commercial batches Stability testing Comparability o Post-approval Manufacturing changes Analytical method changes 5

Analytical method requirements Analytical methods need to be qualified/validated depending on: o Development phase Phase 1 versus commercial o Parameter to be tested Purity, impurities Strength, potency o Intended use In-process control Characterization Release testing Stability testing 6

Validation requirements 1 Type of analytical procedure characteristics IDENTIFICATION TESTING FOR IMPURITIES ASSAY quantitative limit content/ potency Accuracy - + - + Precision Repeatability Interm. Precision - - Specificity + + + + Detection Limit - -/+ + - Quantitation Limit - + - - Linearity - + - + Range - + - + 1: ICH Q2(R1) + + - - + + 7

Validation requirements 2 Validation of analytical procedures during clinical development is seen as an evolving process. For phase I clinical trials, the suitability of the analytical methods used should be confirmed. The acceptance limits and the parameters for performing validation of the analytical methods should be presented in a tabulated form. For phase II and III clinical trials, the suitability of the analytical methods used should be demonstrated. A tabulated summary of the results of the validation carried out should be provided (e.g. results or values found for specificity, linearity, range, accuracy, precision, quantification and detection limit, as appropriate). 2: Guideline on the requirements for quality documentation concerning biological investigational medicinal products in clinical trials (EMA/CHMP/BWP/534898/2008) 8

Validation requirements 3 Proposed acceptable limits supported by simple analytical data, (e.g., IR spectrum to prove the identity, and HPLC chromatograms to support the purity level and impurities profile) of the clinical trials material should be provided Validation data and established specifications ordinarily need not be submitted at the initial stage of drug development. However, for some well characterized, therapeutic biotechnology-derived products, preliminary specifications and additional validation data may be needed in certain circumstances to ensure safety in Phase 1. 3: GfI: Content and Format of Investigational New Drug Application (INDs) for Phase 1 Studies of Drugs, Including Well Characterized, Therapeutic, Biotechnology-derived Products 9

Validation requirements 4 The analytical procedure (e.g., HPLC) used to support the tentative acceptance criteria should be briefly described... [in the IND] A complete description of analytical procedures and appropriate validation data should be available for the analytical procedures that are not from an FDA recognized standard reference... consider the development of stability-indicating analytical procedures that will detect significant changes in the quality of the DS 4: GfI: INDs for Phase 2 and Phase 3 Studies - Chemistry, Manufacturing, and Controls Information 10

Examples - #1 HA: (re: 3.2.S.3.1) The glycosylation profile can influence the pharmacokinetics and/or immunogenicity of the product and so its control should be part of the release testing unless appropriately justified. Company: The applicant agrees to implement a [modification of a characterization method used to characterize N-linked glycans] to assure control of the glycosylation profile in DS. The analytical procedure has been validated and data will be collected for DS batches used in clinical trials to establish acceptance criteria. 11

Examples - #2 HA: (re: 3.2.S.3.1) The complexity of glycosylation of the DS means that a relatively high number of isoforms is present. The applicant should commit to monitoring all DS batches for O-glycan profile and to complete validation of the O-glycan profile method and implement into QC release testing. Company: The applicant proposes to continue O-glycan profiling by extended characterization, by analyzing the next xx full scale DS batches. For QC release testing, an O-glycosylation site occupancy assay will be introduced. 12

Examples - #3 HA: (re: 3.2.P.8) The limit test for detecting oxidized species in stability should be developed and validated as a quantitative method and be implemented as a QC release test for DP. Include resolution between ox peak and main peak as a system suitability criterion. Company: The applicant will implement a QC release test upon validation of the method and include a system suitability criterion (peak-to-valley ratio, USP<621>). 13

Examples - #4 HA: (re: 3.2.S.2.4) In the validation of your HPLC assay, it was stated that the LOD was estimated by dividing the LOQ value by 3. The rationale behind the estimation of LOD was not provided. Provide the reason for the LOD estimated as dividing the LOQ by 3.. Company: Dividing LOQ by 3 is based on the S/N approach for analytical procedures that exhibit baseline noise as outlined in ICH Q2(R1). A S/N of 10:1 is generally acceptable for determination of the LOQ and a S/N between 3 or 2:1 for estimating the LOD is generally acceptable. The multiplication factor between 3:1 and 10:1 is about 3.3. Therefore, LOD was estimated by dividing the LOQ value by factor of 3. 14

Examples - #5 HA: (re: 3.2.P.8) Visible particles should be part of the stability specifications, unless otherwise justified. Company: (Disagreed) We have performed extensive characterization and monitored particles during drug development using visual inspection and micro-flow digital imaging. An extensive stability assessment has also been made including exposure to accelerated, stress, freeze/thaw, and mechanical stress conditions. There was no statistically significant increase in particles and if so, comparable to matching placebo batches. HA: Results acknowledged, however, in the light of previous history with this class of products, importance for patient safety, and limited experience with your specific product, this specification should be included in stability until more experience has been accumulated. 15

Conclusions Regulatory expectations are provided in global guidelines for commerical products, but also for clinical development phases Phase-appropriate method validation is required for in-process tests, release tests and stability tests (comparability) Qualification of methods is required for characterization Health Authorities have asked for characterization tests to be implemented as release tests Health Authorities have asked for a limit test used in stability to become a quantitative test implemented for release testing as well Health Authorities have asked for justification as to the validation approach and to provide this in the dossier Health Authorities have asked for the addition of a test to stability studies. 16

References ICH Guidelines: www.ich.org Guidance for Industry: Analytical Procedures and Methods Validation for Drugs and Biologics Guideline on the requirements for quality documentation concerning biological investigational medicinal products in clinical trials (EMA/CHMP/BWP/534898/2008) Guidance for Industry: Content and Format of Investigational New Drug Application (INDs) for Phase 1 Studies of Drugs, Including WellCharacterized, Therapeutic, Biotechnology-derived Products Guidance for Industry: INDs for Phase 2 and Phase 3 Studies - Chemistry, Manufacturing, and Controls Information 17

Thank you. For questions: info@qualityra.nl