Post-approval Change Management Protocols - Current Status and Next Steps on the Way towards a Global Tool

Similar documents
Application of ICH Q12 Tools and Enablers Post-Approval Lifecycle Management Protocols

ICH Q12 (Pharmaceutical Product Lifecycle Management): PMDA Perspective

Optimising the management of post-approval changes for patients timely access to medicines

Post Approval Change Management Protocols/Comparability Protocols (PACMPs/CPs) Current and Future State

The Ever-increasing Complexity of Biotech Changes - A Pledge for Global Convergence

CMC Forum Europe, 2013

Advantages, Opportunities & Challenges of Adopting the Post Approval Change Management Plan (PACMP)

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

Post-Approval CMC Changes in Japan: How We Envision the Future

Current Status and Perspectives on Pharmaceutical Products in Japan

THE PACMP STRATEGY. March 13-16, 2018

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

CASSS CMC Strategy Forum EU Introduction to EBE Satellite Session

PDA & West Present: Combination Products Combination Product Hot Topics: Post Approval Device Changes and the New EU MDR Article 117 Requirements

QbD Approach and Regulatory Challenges in Japan

Advanced Concepts for Change Control of Analytical Procedures with ICH Q12 Coming

Introduction and Update on ICH Q12 Guideline Dr Frank Montgomery, Global Head, Reg CMC, AstraZeneca/Medimmune Q12 EFPIA Expert

Update on the Regulatory Considerations of Pharmaceutical Product Lifecycle Management (ICH Q12) from a Health Authority Perspective

ICH Q12 : A Unique Opportunity to Realize 21st Century Quality Vision

PMDA Perspectives Yoshihiro Matsuda, Ph.D.

EU Update on Regulatory Developments

Current Hotspots during CMC Evaluation a European Regulatory Perspective

Linking Regulatory Commitments to Post Approval Changes Robert Iser

CMC Consideration for the Development of Regenerative Medical Products

Post-approval Variations

Will ICH Q12 truly goes beyond Q8/11?? Opportunities and Challenges

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

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

Post Approval Changes & Product Lifecycle Management

PMDA Perspective: Recent Trends in the Regulation of Biopharmaceuticals

Post approval change of Japanese registration dossiers and impact on market supply

TECHNICAL AND REGULATORY ASPECTS OF PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT: ICH Q12

Experience with Health Canada s Approach for Post-Approval Changes. Kiran Krishnan Vice President US Regulatory Affairs September 2014

PPTA Regulatory Workshop June 13, 2016

Impact factor: 3.958/ICV: 4.10 ISSN:

While the recognition

ICH Q12 Technical and Regulatory Considerations for Pharmaceutical Product Lifecycle Management Møde i Industrifarmaceut foreningen 15/

An Industry Perspective: Reducing the Complexity and Impact of Regulatory Changes in Latin America

Quality (or CMC) Enablers for Efficient/Effective Drug Product Lifecycle Management FDA s Perspective

A Late-Stage Monoclonal Antibody in the FDA QbD Pilot Program: Moving from Concepts to Implementation

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

Introduction to CMC Regulatory Affairs

Quality Risk Management and Submission Strategies for Breakthrough Therapies

Challenges and Opportunities in the Development of integrated Drug Device Combination Products CMC Strategy Forum, Tokyo, 5 December 2017

Challenges in Getting Global Approvals for Post-approval Changes

ABB Industries PAT Validation

The Device Side of Combination Products

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

ICH Q12 Technical and Regulatory Considerations for Pharmaceutical Product Lifecycle Management

TECHNICAL AND REGULATORY CONSIDERATIONS FOR PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT Q12

COMMENTS FROM EUROPABIO GENERAL COMMENTS

Regulations for Post Approval Changes of Biotherapeutics Needs and Opportunities

Lessons Learned from QbD Application of Biologics in Japan: Perjeta Case

Application of ICH Q12 Tools and Enablers

Quality by Design: An Attempt to Jumpstart. Peter Calcott, Ph.D. President, Calcott Consulting

Replacing Analytical Methods for Release and Stability Testing CBER Perspective

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

TECHNICAL AND REGULATORY CONSIDERATIONS FOR PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT Q12

Status of the ICH Q11 Guideline on Development and Manufacture of the Active Substance. Riccardo Luigetti Prague, 9 December 2009

TECHNICAL AND REGULATORY CONSIDERATIONS FOR PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT Q12

An Industry Perspective: The Complexity of Postapproval CMC Changes and Proposed Regulatory Strategies. SPEAKER: Suzanne Murray Biogen

Biotechnology Industry Organization 1225 Eye Street NW, Suite 400 Washington, DC 20006

THE NEW QUALITY PARADIGM OPPORTUNITIES AND EXPECTATIONS IN ICH Q8 Q9 Q10 Q11 DR. FRITZ ERNI

Introductions and Perspectives on International Harmonization

Products: A Global Perspective

A holistic regulatory approach to accelerated CMC development

Themes & Key Points The Problem Statement

Impacto de las Nuevas Tendencias Regulatorias en la Producción de Parenterales. Innovacion en Packaging Primario

ICH Quality Implementation Working Group POINTS TO CONSIDER

Revision of the Variations Regulation. Key Principles and Guidelines Development CONTENT

PhUSE Single Day Event 2009, Frankfurt

Established Conditions: Reportable CMC Changes for Approved Drug and Biologic Products Guidance for Industry

Setting the Scene CHALLENGES & COMPLEXITIES OF POST-APPROVAL CMC / LABELING CHANGES

Implement an Effective Change Management System throughout GMP and Validation Environments. Eileen Cortes February 23, 2017

Future of Question-based Review and Regulatory Submissions

Change Control Overview

CMC Workshop CMC Challenges in Japan. April Bethesda, MD. Yoshihiro Matsuda, Ph.D., PMDA Session 10

PROCESS VALIDATION ROCHAPON WACHAROTAYANKUN, PH.D.

EU Update on Regulatory Developments

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

QbD approach and Regulatory Challenges in Europe

Variation Regulations (EU)

Regulation of Active Pharmaceutical Ingredients (API)

PMDA Perspective: Regulatory Updates on Process Validation Standard

Setting Specifications for Biotech Products

The Application of Pharmaceutical cgmp to Live Bacterial Products. James Harris Business Development Manager

Replacing Analytical Methods for Release and Stability Testing A Regulatory Perspective

Welcome to the CMC Strategy Forum Europe 2016

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

Regulatory Updates in Taiwan

ICH Q11 Development & manufacture of drug substances

Welcome to the CMC Strategy Forum

Guidelines. of

Planning of Application

Regulatory Updates for Biopharmaceutical Products:FDA Perspective

Official Journal C 223. of the European Union. Information and Notices. Information. Volume 56 2 August English edition

Welcome to CMC Strategy Forum Japan 2015

Global Biologics Regulatory Trends Challenges and Opportunities..

PPQ-to-Approval Timelines Acceleration Approaches at BMS

Transcription:

Post-approval Change Management Protocols - Current Status and Next Steps on the Way towards a Global Tool Dr. Markus Goese Lead EU CMC Regulatory Policy F. Hoffmann-La Roche Ltd, Basel - Switzerland

Presentation outline What is a PACMP? What are the benefits of protocols? What are the challenges? What is a CP? Expansion of the protocol concept (ecp) The Global context & way forward in ICH Q12 2

Question-1 So what is a PACMP and what can be done with it? 3

Change Management Protocols in EU Post-approval change management protocol (PACMP) concept introduced in 2010 in EU through Variations Classification: 4

What is a PACMP? A PACMP describes specific change(s) that a company would like to implement following marketing authorization and how these would be prepared and verified A PACMP applies to all types of products and incorporates a science and risk-based approach to evaluate impact of change(s) on product quality in a proactive manner PACMPs may be included in an original marketing authorization application (MAA) or be submitted as a stand-alone type II-variation; approved PACMPs can be modified via a type II (major change) or type IB (minor change) variation 5

Reminder: Variation categories in the EU Most variations for biologics are major (type II) changes Category Impact on quality, safety or efficacy? Notification and implementation Review time Type IA None / minimal impact Notification: within 12 months after implementation 30 days Type IA IN None / minimal impact Notification: immediate following implementation 30 days Type IB Minor potential impact; Classified by default, if change does not fall into another category Notification: before implementation, but no formal approval Implementation: tell, wait and do MAH implements changes unless negative opinion received within 30 days 30 days Type II Major potential impact Notification: EMA forwards positive CHMP opinion to EU Commission, and they amend the MA within one year Implementation: following CHMP opinion, without waiting for Commission approval 60 days normally 90 days for indication changes 30 day for urgent safety issues 6

The principle of the Change Management Protocol: a 2-step implementation approach «traditional» approach without PACMP: with PACMP: Source: EMA Questions and Answers on post-approval change management protocols 7

The Contents of a Post-Approval CMP, 1/2 According to EMA PACMP Q&A: Justification that there is a recognized future need for the specific change within a reasonable timeframe. A detailed description of the proposed change. Risk assessment of the impact of the change on product quality. Discussion on the appropriateness of the approved control strategy to identify and manage these risks. 8

The Contents of a Post-Approval CMP, 2/2 Description of the studies to be performed, the test methods and acceptance criteria that will be used to fully assess the effect of the proposed change on product quality. For biologics, the approach to be used to demonstrate the comparability of the pre-, and post-change product. A plan for stability studies should be included, if appropriate. In case that the protocol describes several changes, a justification showing that the changes are related, and that a simultaneous review under a single protocol is meaningful. For small molecules, a proposed reporting category for step 2 (type IA or IB variation). 9

EU Experience: PACMPs authorised in the centralised procedure (last updated 22/05/2015, source: EMA) SCOPE TYPE II MAA Line extension New site for manufacture and/or QC testing of the drug substance Bio: 12 Bio: 1 Che: 4 New site for manufacture and/or QC testing of the drug product Bio: 18 Bio: 3 Che: 2 Change to the manufacturing process of the drug substance Bio: 12 Bio: 2 Che: 1 Scale-up of the drug substance manufacturing process Bio: 3 Bio: 2 Change to the preparation of a cell bank Bio: 1 Bio: 2 Change to the manufacturing process of the drug product Bio: 2 Che: 1 Change to the container closure system of the drug substance or drug product Bio: 1 Che: 1 Other Bio: 1 TOTAL - 60 for biologics: 10 MAA, 49 Type II, 1 Extension - 9 for chemicals: 7 MAA, 2 Type II Note: 4 PACMPs for biologics not included in the table were withdrawn (2 MAA, 2 Type II) Limited use so far for chemical Drug Substance/ Drug Product 10

Why use a PACMP approach? Benefits: Expedited review and/or inspection at step 2 of PACMP procedure Reduced category for future reporting of CMC changes covered by the approved protocol (but type IB default for biologics in EU) Predictability and transparency in terms of requirements and studies needed to implement a change (approved protocol is an agreement between the sponsor and the HA) Faster implementation, if the pre-determined criteria of the PACMP are met; use of the PACMP could allow an applicant to implement CMC changes and place a product in distribution sooner than without the use of the PACMP procedure Could also prove to be very useful regulatory tool in breakthrough/ accelerated pathway scenarios, to supplement specific parts of initial dossier in a pre-agreed manner. 11

Example (EU): Biologics DS manufacturing site transfer - Benefit of PACMP Approach vs. Traditional Approach* 3-5 months faster approval of the site change using a PACMP *Note: approval timelines for type II variation in this scheme include positive CHMP opinion and Commission Decision 12

Some specific considerations for PACMPs The types of changes that could be included in a protocol depend on the complexity of the product and its manufacturing process, as well as the understanding that the company has gained about them. For a (biological) medicinal product where non-clinical/ clinical data are needed as part of the comparability exercise, a post approval management protocol will not be feasible. Change Management protocols are applicable to all types of applications, irrespective of the development approach that has been followed, i.e., traditional or enhanced; However, it is expected that if the applicant has applied QbD principles during product development, then an increased product and process understanding is achieved, thus making it easier to predict the impact of a change on the active substance or finished product quality. 13

Question-2 And what is a CP? 14

Comparability Protocols in the US FDA issued Draft Guidance for Industry Comparability Protocols - Chemistry, Manufacturing, and Controls in 2003 Revision is on the FDA agenda of planned guidance to be published in 2015 Overall, EU-PACMP and US-CP concept very similar 15

Reminder: US Post-Approval Change Reporting Categories Submission Type Prior Approval Supplement (PAS) Changes Being Effected, 30 days (CBE-30) Changes Being Effected (CBE) Annual Report (AR) Potential impact Product may be distributed FDA Approval Timeline Substantial After approval 4 months Moderate Moderate 30 days after FDA receipt Immediately following FDA receipt 6 months 6 months Minimal No restrictions 180 Days First step of CP submitted with NDA/ BLA or as Prior Approval Supplement (PAS), second step allows for reduced reporting category if predefined criteria are met (e.g., CBE-30) 16

Example (US): Manufacturing Site Transfer of Product A from Site X to Site Y Leveraging traditional CP Defined business need Submit CP describing site transfer acceptance criteria for productsite combination A B C Product A Execute transfer per defined requirements in CP D A CBE-30 Supplement with data demonstrating acceptance criteria met Drug Substance Donor Site X Drug Substance Receiving Site Y Time savings for US-CP-approach comparable to EU- PACMP, i.e. up to 5 months for site-transfer (product release to market) 17 17

Question-3 CP + QbD Concepts =? 18

Question-3 CP + QbD Concepts =? CP + QbD Concepts = Expanded Comparability Protocol (ecp) 19

Expanded Comparability Protocol (ecp) Term ecp was first introduced at the US CASSS CMC Forum in July 2009 on QbD as concept for how to manage lifecycle changes based on risk ecp leverages existing regulations for CPs: move from one protocol for one change to an expanded protocol applicable to groups of similar changes (across products) A range of QbD concepts (ICH Q8, Q9, Q10, Q11) can be applied: leverages historical knowledge, systematic risk assessment and change requirements Genentech s drug substance manufacturing site transfer ecp proposal was accepted in FDA s QbD pilot program in 2009 Primary objective of the site transfer ecp: support the mobility of approved products to licensed drug substance facilities within Roche/Genentech s manufacturing network ( trans-bla approach : multiple products/ multiple sites) Effectively leverages Quality Risk Management (ICH Q9) and builds upon company s knowledge/ experience with site transfers across products & reduces number of regulatory submissions of similar content, drives consistency 20 20

Concept of ecp to Support Multi-Product, Multi-Site Transfers A network of Drug Substance Donor Sites A Site X Site Z Potential Future Network Requirements A B A network of Drug Substance Receiving Sites Site Y or Z Site X or Z SiteY D C Site X or Z B C D Site X or Y Submit ecp (PAS) describing acceptance criteria broadly for both Site and Product Execute transfer per defined requirements in ecp 21 CBE-30 Supplement with data demonstrating acceptance criteria met 21

Design of Site Transfer ecp Risk Assessment Tools ecp CBE-30 Site Inspection Comparability & Validation GMP/Compliance Product and Process Evaluation Important questions to be asked: What should be assessed in considering GMP or inspectional risk? What should be tested to confirm comparability? Site Transfer Risk Assessment/ Quality Risk Management Scope and Limitations Facility modifications What are the impacts of facility fit changes? Risk-Based Approach to What products and process/facility facilities should be in scope? 22 22

Snapshot: Site Transfer Risk Assessment Failure modes prospectively identified and assessed by team of experts Note: Risk Priority Number (RPN) is the product of the Primary Risk Number (PRN) and the score for probability of detection. Subsequent step: Risk Prioritization - Identified thresholds of risk acceptability incorporated in overall applicability to Site Transfer ecp 23 23

Multi-Site, Multi-Product ecp approved in FDA QbD Pilot Program for Biotech Products Main points accomplished: Agreed upon scope: products and facilities (scope excludes opportunistic process changes including changes to increase yield; included within scope: facility driven process modifications, eg. scale changes, disposables, raw material sourcing) Justified that historical knowledge of product characterization, stability & process performance is sufficient to support comparability with post-approval real-time stability commitment (successful demonstration of comparability using stress stability studies) Only licensed facilities in a current state of GMP compliance are allowed, so that Pre-Approval Inspection (PAI) may be waived Agreed future products and facilities could cross-reference the ecp if pre-defined criteria were approved Overall: ability to quantify risk related to facility and process change: ecp PAS approved in 2011, Subsequent drug substance transfers approved under ecp criteria with CBE-30 24 24

The Global context & way forward with ICH Q12 25

The Global context & way forward for PACMPs with ICH Q12 It is planned that the upcoming ICH Q12 guideline contains a chapter describing post-approval change management protocols as an important tool for efficient lifecycle management Based on current FDA and EMA guidance (showing a high degree of similarity), first draft of this chapter has been discussed & agreed during the Q12 EWG meeting in Fukuoka high-level contents is very much in line with the PACMP/ CPprinciples, need for wider use & broader protocols confirmed Swissmedic has PACMPs already in place PMDA/ MHLW have indicated their willingness to adopt the concept in Japan excellent foundation for protocol harmonization efforts under the lead of ICH 26

«Last note» on Q12: Lifecycle strategy & PACMPs The application file should contain a section summarizing the essential control strategy elements (at time of submission) learned from the science and risk-based pharmaceutical product development. In addition, for all new registration dossiers, this section should also include a subsection on lifecycle management strategy (future planned changes) derived from the information/data submitted in the original application file (dossier). [ ] The section includes: References to any post-approval change management protocols (PACMPs) contained within the dossier [ ] These protocols serve as main regulatory tool that describe future changes to the Established Conditions, the potential impact and reporting category of the change as well as the underlying data requirements for implementation 27

to be continued @ next ICH meeting www.visitjacksonville.com

Acknowledgements - Julia Edwards, Biogen - Wassim Nashabeh, Roche - Pascal Venneugues, EMA - Jutta Riedl, Swissmedic - Jean-Louis Robert, EMA/QWP - Ashley Boam, Bob Iser; FDA/CDER - Ingrid Markovic; FDA/CBER - Yasuhiro Kishioka, PMDA - Pierrette Zorzi, consultant 29

THANK YOU! -- Questions? http://kyushu-japan-holidays.com/ 30

Backups 31

Protocols in US-Regulation (BLA) 21 CFR 601.12(e) An applicant may submit one or more protocols describing the specific tests and validation studies and acceptable limits to be achieved to demonstrate the lack of adverse effect for specified types of manufacturing changes on the identity, strength, quality, purity, or potency of the product as they may relate to the safety or effectiveness of the product. Any such protocols, or change to a protocol, shall be submitted as a supplement requiring approval from FDA prior to distribution of the product which, if approved, may justify a reduced reporting category for the particular change because the use of the protocol for that type of change reduces the potential risk of an adverse effect. 32

Doing now what patients need next