Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage.

Slides:



Advertisements
Similar presentations
Strengthening the Medical Device Clinical Trial Enterprise
Advertisements

1 Implementation of Quality by Design (QbD): Status, Challenges and Next Steps Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS,
VALIDATION What is the new guidance?. What is a Compliance Policy Guide? Explain FDA policy on regulatory issues CGMP regulations and application commitments.
Integrating CMC Review & Inspection Industry Recommendations Joe Anisko April 24, 2003.
Determine impurity level in relevant batches1
Implementation of Quality-by-Design: ONDQA Initiatives Advisory Committee for Pharmaceutical Science October 5, 2006 Chi-wan Chen, Ph.D. Deputy Director.
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.
Manufacturing Subcommittee of the Advisory Committee for Pharmaceutical Science July 20-21, 2004 Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.
COMPARABILITY PROTOCOLS ACPS March 12-13, 2003 Stephen K. Moore, Ph.D. Chemistry Team Leader CDER/Office of New Drug Chemistry Co-Chair, Comparability.
Pilot Risk-Ranking Model to Prioritize Manufacturing Sites for GMP Inspections Advisory Committee for Pharmaceutical Science Manufacturing Subcommittee.
Assessing Quality-by-Design A CMC Review Perspective
Training Workshop on Pharmaceutical Development with a Focus on Paediatric Medicines / October |1 | Regulatory Requirement on Dossier of Medicinal.
Challenges and Opportunities in Enhancement of the CMC Section of NDAs: Quality – by - Design Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.
Individual Bioequivalence Lawrence J. Lesko, Ph.D. Director Office of Clinical Pharmacology and Biopharmaceutics Advisory Committee for Pharmaceutical.
PAT Validation Working Group Process and Analytical Validation Working Group Arthur H. Kibbe, Ph.D. Chair June 13, 2002.
ONDQA Perspective on Post Approval Changes Eric P. Duffy, PhD Director, Division of Post-Market Evaluation, ONDQA, CDER, FDA Public Meeting: Supplements.
1 Revisions to 21 CFR Supplements and Other Changes to an Approved Application PhRMA Perspective FDA Public Meeting – 7 Feb 2007.
Learnings from Pre-approval Joint Inspection of a GSK QbD Product with US-FDA & EMA and the application of Continuous Verification 17 May 2011, Beijing,
Good Clinical Practice GCP
Ensuring Physical Stability of Pharmaceuticals: Can/should we improve our ability to identify and prevent physical changes? Ajaz S. Hussain, Ph.D. Deputy.
Application of the principles of QbD in vaccines production Andrea Pranti.
Risk-Based CMC Review Paradigm
Organizational Gaps in Reaching the “Desired State” Helen Winkle.
Slide 1 May 2008 Training Workshop on Pharmaceutical Development with focus on Paediatric Formulations Mumbai, India Date: May 2008 QUALITY BY DESIGN.
Performance Measurement and Analysis for Health Organizations
Quality by Design Application of Pharmaceutical QbD for Enhancement of the Solubility and Dissolution of a Class II BCS Drug using Polymeric Surfactants.
Establishing Drug release/Dissolution Specifications – QBD Approach Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS, CDER Advisory.
Excipient QbD Concepts to Enhance the Development of Robust Drug Products Priscilla S. Zawislak Global Regulatory Affairs Manager - Ashland Inc. Chair.
1 Supplements and Other Changes to an Approved Application By: Richard J. Stec Jr., Ph.D. February 7, 2007.
FDA’s Advisory Committee for Pharmaceutical Science The Subcommittee on Process Analytical Technologies (PAT): Overview and Objectives Ajaz S. Hussain,
Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Science, CDER, FDA ACPS Subcommittee on Manufacturing Science: Identification and Prioritization.
Introduction to Topic #1: QbD approach for quality control and assurance of Dissolution Rate Ajaz S. Hussain, Ph.D. Deputy Director, Office of Pharmaceutical.
Quality by Design (QbD) Myth : An expensive development tool ! Fact : A tool that makes product development and commercial scale manufacturing simple !
1-7.The ICH Q8 “Minimal Approach” to Pharmaceutical Development
1 Regulatory Aspects of Pharmaceutical Excipients PQRI Workshop Nick Buhay Acting Director Division of Manufacturing and Product Quality Office of Compliance.
1 An Update on ICH Guideline Q8 – Pharmaceutical Development FDA Advisory Committee for Pharmaceutical Science: 5 Oct 2006 Dr John C Berridge Senior Regulatory.
Proposal for End-of-Phase 2A (EOP2A) Meetings Advisory Committee for Pharmaceutical Sciences Clinical Pharmacology Subcommittee November 17-18, 2003 Lawrence.
Quality by Design & Question-Based Review: Observations by the Generic Pharmaceutical Industry Advisory Committee for Pharmaceutical Science October 5,
Overview of FDA's Regulatory Framework for PET Drugs
Challenges and Opportunities in Enhancement of the CMC Section of NDAs: Quality – by - Design Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical.
1 Basis of the Proposed Tactical Plan for a QbD approach for Quality Control and Assurance of Dissolution Rate Ajaz S. Hussain, Ph.D. Deputy Director,
What Impact should ICH Q8 have on ICH Q6A Decision Trees?
PhRMA Perspective on FDA Final Report FDA Advisory Committee on Pharmaceutical Sciences October 20, 2004 G.P. Migliaccio, Pfizer Inc.
Risk-Based CMC Review - OGD Perspective Gary J. Buehler, R.Ph. Director Office of Generic Drugs July 21, 2004 Advisory Committee for Pharmaceutical Science.
Molecule-to-Market-Place Quality
COMPARABILITY PROTOCOLUPDATE ADVISORY COMMITTEE FOR PHARMACEUTICAL SCIENCE Manufacturing Subcommittee July 20-21, 2004 Stephen Moore, Ph.D. Chemistry Team.
Bioequivalence of Locally Acting Gastrointestinal Drugs: An Overview
10:00 A.M. – Noon 7 June 2004 ICH Quality Plenary Meeting.
Drug Release Specification: In Vivo Relevance Ajaz S. Hussain, Ph.D. Deputy Director, OPS/CDER/FDA.
Progress in FDA’s Drug Product Quality Initiative Janet Woodcock, M.D. November 13, 2003.
1 Office of Pharmaceutical Science on Jon Clark FDA/CDER/OPS Associate Director for Policy Development.
General Aspects of Quality assessment of multisource interchangeable medicines Rutendo Kuwana Technical Officer, WHO, Geneva Training workshop: Assessment.
Using Product Development Information to Address the Bioequivalence Challenges of Highly-variable Drugs Lawrence X. Yu, Ph. D. Director for Science Office.
Pharmaceutical Manufacturing Subcommittee of the ACPS Ajaz S. Hussain, Ph.D. ACPS Meeting October 22, 2002.
Bioavailability of Dietary Supplements: Key Issues in Defining the Research Agenda Impact of Formulation on Bioavailability? Discussion Leader: Stephen.
CDER / Office of Compliance ACPS October 5, 2006 Joseph C. Famulare Acting Deputy Director Office of Compliance CDER / FDA.
Second Meeting of the FDA/ACPS Process Analytical Technology: Closing Remarks Ajaz S. Hussain, Ph.D. Deputy Director Office of Pharmaceutical Sciences.
FDA’s Advisory Committee for Pharmaceutical Science The Subcommittee on Process Analytical Technologies (PAT): Closing Remarks Ajaz S. Hussain, Ph.D. Deputy.
Bioequivalence Criteria Research Plan Stella G. Machado, Ph.D. Office of Biostatistics and the Replicate Design Technical Committee Advisory Committee.
Examining Drug Quality Regulation Douglas C. Throckmorton, MD Deputy Director Center for Drug Evaluation and Research Public Meeting on 21 CFR February,
Topic #2: Quality by Design and Pharmaceutical Equivalence Ajaz S. Hussain, Ph.D. Office of Pharmaceutical Science Center for Drug Evaluation and Research.
开发报批美国 FDA 的仿制药 与相关问题探讨 上海复星普适医药科技有限公司何平. 内容提要 开发仿制药的重要性和机遇 开发仿制药的重要性和机遇 开发仿制药的挑战 开发仿制药的挑战 申报仿制药的分类 申报仿制药的分类 仿制药研发团队 仿制药研发团队 仿制药的研发过程 仿制药的研发过程 QbD 在制剂开发中怎么体现.
Product & Process Working Group February 26, 2002.
In vitro - In vivo Correlation
Michael Kopcha, Ph.D., R.Ph. Director Office of Pharmaceutical Quality
An Update on ICH Guideline – Pharmaceutical Development
Deputy Director, Division of Biostatistics No Conflict of Interest
Quality System.
QUALITY BY DESIGN Training Workshop on Pharmaceutical Development with focus on Paediatric Formulations Mumbai, India Date: May 2008.
Implementation of Quality by Design (QbD): Status, Challenges and Next Steps Moheb M. Nasr, Ph.D. Office of New Drug Quality Assessment (ONDQA), OPS, CDER.
Presentation transcript:

Achieving and Demonstrating “Quality-by-Design” with Respect to Drug Release/dissolution Performance for Conventional or Immediate Release Solid Oral Dosage Forms Ajaz S. Hussain, Ph.D. Deputy Director, OPS/CDER/FDA FDA’s ACPS Meeting October 2005

Topic Introduction At the May 2005 meeting of the ACPS we proposed that significant opportunities exist to further improve the effectiveness and efficiency of dissolution rate control and related regulatory decisions: –FDA’s PAT & CGMP Initiatives; the shared vision (“desired state”) ICH Q8 Guideline and PAT Guidance –PQAS [ONDQA, QbR in OGD,…] –FDA proposed tactical plan

Topic Introduction At the May 2005 meeting we extended invitations to all stakeholders to consider our proposed tactical plan as a first step and to comment and/or develop their proposals. –Today, USP, GPhA, and PhRMA will present their perspectives/proposals –A report on dissolution test variability from two academicians (included in the background packet) –FDA presentations

QbD Guiding Principles? The term “quality-by-design” has been the foundation of the current regulatory system; yet there is a lack of common understanding or uncertainty of what this means –High degree of variability on how different companies approach quality-by-design –QbD approach to drug release specification can serve to illustrate the fundamental guiding principles

QbD Guiding Principles: Drug Release Rate Rate of drug release from solid oral dosage forms is a critical quality attribute A desired release characteristic (in vivo, target value and acceptable variability) should be designed to meet the performance objectives (intended use) of a proposed product in the intended patient population - Design specification

QbD Guiding Principles: Drug Release Rate Ideally, design specifications should be proposed and established early in drug development such that the pivotal clinical trial product produced is in conformance. For conventional dosage forms, certain design specifications, or certain aspects of specifications, are generally based on prior knowledge. –Bioequivalence goal post (90% CI T/R in % range); IR dosage forms: Q-15%, extended release dosage forms: Q ± 10%

QbD Guiding Principles: Drug Release Rate Design specification decisions should be guided by information obtained from pre-clinical and pre-formulation drug characterization Design specifications then guide the development of a proposed product, its manufacturing process, and the quality assurance strategy Structured product and process development should identify a set of variables and their ranges that can reliably deliver the desired design specification (Design space)

QbD Guiding Principles: Drug Release Rate Clinical evaluation during various phases of drug development provides both quantitative (e.g., pharmacokinetics/pharmacodynamics analysis) and qualitative (e.g., clinical observations) opportunities to verify that selected design specifications were achieved and are optimal for the intended use. –These opportunities should be leveraged as early as feasible to maximize the likelihood that a product design can be used in pivotal clinical trials and can be considered to have achieved quality-by-design (e.g., design specification and control strategy).

QbD Guiding Principles: Drug Release Rate When regulatory evaluation of the clinical safety and efficacy reaches a conclusion to approve a new drug application: –clinical trial product design specifications should become regulatory specifications, and –a regulatory risk-based control strategy is established to ensure that production lots will consistently deliver a set of regulatory- design specifications

QbD Guiding Principles: Drug Release Rate The inherent variability (“common cause”) in a clinical trial product design is qualified through –the structured product and process development information that demonstrates that critical variables relevant to a product and process design were identified and adequately controlled (i.e., all significant “special cause” variability), and – acceptable performance of the product clinical trials

QbD Guiding Principles: Drug Release Rate It is recognized that during drug development, a limited number of batches are generally manufactured and the scale of manufacture may be smaller compared to the “to-be-marketed” batches –In a QbD paradigm, limited manufacturing experience should not impact on design specifications, but it can be related to the establishment of “alert” and “action” process and product control limits by a sponsor

QbD Guiding Principles: Drug Release Rate Following scale-up and/or technology transfer (R&D to Industrial Operations) the “action” and “alert” limits may need to be modified to ensure that a process remains in a desired state of control –These decisions should be based on sound scientific basis and the principles of statistical process control Managed under a company’s quality system and subject to CGMP regulatory inspections

QbD Guiding Principles: Drug Release Rate Product and process control strategy should be designed to facilitate continuous quality verification (as opposed to discrete “3 batch – process validation”) and continuous improvement (e.g., improving efficiency, reducing variability,…) –Regulatory - design specifications should be articulated in terms of “continuous variables” as opposed to discrete counts (e.g., no unit is outside..) –Understanding the sources of variability and measuring and controlling critical material attributes (raw and in-process materials) as a means for process control Minimize the need for using process “time” and machine settings as the primary means for process control –Incorporating engineering control approaches as opposed to primary reliance on end-product testing after a batch has been manufactured

QbD Guiding Principles: Drug Release Rate Dissolution testing is a tool for: –Product development & optimization –Quality control –Product characterization and comparison for decisions of waiver of in vivo bio studies –Establishing performance tests in compendial monographs A clear distinction of the purpose for which this tool is to be used is necessary –E.g., a part of the quality control strategy vs. waiver of in vivo bio studies

QbD of Clinical Trial Product “Design Specifications & Control Strategy” Clinical Studies Acceptable Safety & Efficacy Demonstrated in Clinical Trials Pre-Clinical Studies Pre-Formulation Studies Prior Knowledge (Manufacturing Science) Regulatory Specifications and Control Strategy Risk Based CGMP Inspections & Continuous Improvement Post-Approval AERS & Risk Mgmt Summary: QbD Flow Diagram