GPhA Fall Technical Conference Nov 2-5, 2015 Bethesda, MD ICH M7 Guidance Overview and Current FDA Perspectives

Similar documents
Starting Material Selection for Type II Drug Master Files

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

Regulatory Starting Materials An FDA Perspective

Drug Substance Review in the Office of Pharmaceutical Quality

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

Introduction to CMC Regulatory Affairs

Regulatory Review Considerations of Drug-Linker Quality in ADCs

API Selection of Starting Materials Impact on First Cycle Approval

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

API Testing Requirements to Support the EI Risk Assessment. Elisabeth Corbett Associate Director, GRS-CMC, Bristol-Myers Squibb November 9, 2016

Guidance for Industry

Flexible and Pending Monographs

BRIEFING. . Over time, 466 may be used less frequently and may be withdrawn.

Early Development Best Practices for Stability- Regulatory Perspective

Strategic Implantation of PAT : FDA Perspective

S9 Implementation Working Group ICH S9 Guideline: Nonclinical Evaluation for Anticancer Pharmaceuticals Questions and Answers

Bench to Bedside - The Roadmap Chemistry, Manufacturing, and Controls Issues in Radiopharmaceutical Applications

New Drug Product Impurities

Pharmaceutical Development (Drug Substance & Drug Product) for Visceral Leishmaniasis candidate DNDI-6148

Further Stability Considerations

ICH Q11 Development & manufacture of drug substances

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

Guidelines: c. Providing the investigational drug for the requested use will not interfere with the initiation, conduct, or completion of clinical

Examples of regulatory expectations for analytical characterization and testing

IMPURITIES. Antony Fake API Focal Point, PQTm WHO PREQUALIFICATION TEAM MEDICINES

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

Hot Topics in Drug Product Process Validation: A Reviewer s Perspective

Setting Specifications for Biotech Products

IMPURITIES IN NEW DRUG PRODUCTS

Best Practices and Application of GMPs for Small Molecule Drugs in Early Development IQ Workshop, Feb 4-5, 2014, Washington, D.C.

Derivation and Justification of Safety Thresholds

WHO GUIDELINE Stability testing of active pharmaceutical ingredients and finished pharmaceutical products

The Role of Chemists in the FDA Drug Approval Process

How to Identify Critical Quality Attributes and Critical Process Parameters

FDA approval of emergency expanded access use may be requested by telephone, facsimile, or other means of electronic communications.

Document Reuse: Theory and Practice

QbD (Quality by Design) Has industry benefited from this? WHITE PAPER.

Clinically Relevant Dissolution Specifications: FDA Perspective and Initiatives

COMMERCIAL PRODUCT STABILITY

Regulatory Considerations and Trends Europe and the U.S.

Analytical Methods Development and Validation

TECHNICAL AND REGULATORY CONSIDERATIONS FOR PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT Q12

Evolution of the CMC Review - ANDAs

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

USP Chapter 823 USP 32 (old) vs. USP 35 (new)

Method Validation by Phase of Development

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON THE NON-CLINICAL DEVELOPMENT OF FIXED COMBINATIONS OF MEDICINAL PRODUCTS

Drug Development Services

ISPE S GUIDES AND HOW THEY APPLY TO CLEANING AND CLEANING VALIDATION

Extractables and leachables: An Introduction

Replacing Analytical Methods for Release and Stability Testing CBER Perspective

In silico strategies to assess potentially mutagenic impurities under ICH M7

Balancing the time, cost and risk of drug development. Christina Gustafsson, PhD Pharm, Formulation Scientist at Pharmaceutical Development, APL

Case study 2: Parenteral Drug Product

PhUSE Single Day Event 2009, Frankfurt

GUIDANCE FOR INDUSTRY ON FIXED DOSE COMBINATIONS (FDCs)

Structure and content of an IMPD. What is required for first into man trial?

QbD Concepts Applied to Qualification and Transfer of Analytical Methods

Bioavailability and Bioequivalence Studies

MASTER FILE PROCEDURES

Industry Experience: Early Collaboration with FDA on Combination Products. Kristi Kistner, Amgen Inc. CMC Strategy Form January 26, 2015

Copyright. Jeremiah J. Kelly (2015). All rights reserved. Further dissemination without express written consent strictly prohibited.

GUIDELINE FOR THE STABILITY TESTING

CEP submission: How to prepare a New Application?

KFDA Regulatory Framework on Biopharmaceuticals - Focus on Biosimilar

October 10, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm Rockville, MD 20852

Challenges in Implementing Knowledge Management within ICH Q10

Guidance Document 01 January 2016 CONTENTS. 1. Introduction Background 1.2. Objectives 1.3. Scope and application 1.4 APIMF holder obligations

The Drug Development Process and Design of Clinical Trials

ICH Q3D Guideline Impact on the Users: Perspective of a Finished Product Manufacturer John Glennon

GUIDANCE FOR QUALITY ASSESSORS DRUG SUBSTANCE. IGDRP Quality Working Group

Systematic Risk Management: An Overview of ICH Q-9

CTA/NDA Regulatory Landscape in China. Jack Xie, PhD, DABT SOT 2016

Pharma EXPO 6 9 November 2016 Chicago, IL

Supply of IND Agents to Multi-center trials by Skilled Academic Sites

September 12, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852

LEGAL REQUIREMENTS FOR STABILITY

Investigating OOS for Finished Product on the Stability Program. Presented by: Nicole Chang, QA Manager, Apotex Pty Ltd


The Device Side of Combination Products

The Investigator s Brochure: A multidisciplinary document

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

Demonstrating a High Degree of Assurance in Stage 2 of the Process Validation Lifecycle

US FDA: CMC Issues for INDs

Regulatory perspectives on CQAs, CPPs, and Risk Analyses for Combination Products.

Outline CLINICALLY RELEVANT SPECIFICATIONS. ISPE Process Validation Conference September 2017 Bethesda, MD

Overview of FDA Regulations and Guidance Documents related to PET Drug Chemistry. Steve Zigler, Ph.D. Siemens PETNET Solutions

Leachable and Extractable Testing

The Emerging Technology Program: FDA s Perspective

International Transfers of Personal Data at sanofi-aventis R & D

Office for Human Subject Protection. University of Rochester

European Medicines Agency Evaluation of Medicines for Human Use

Continuous Manufacturing Achieving the Vision of Modernizing Pharmaceutical Manufacturing

Draft regional guidelines on stability testing of active substances and pharmaceutical products

BIOPHARMA SOLUTIONS TM Expedite Your Drug Development Program

PHARMACEUTICAL TESTING

Exclusive Manufacturing Solutions

Optimization Strategies of Expression Cell Line Construction to Reduce the Biological Drug Development Risk. Feng Gao, MD, PhD. AutekBio CO.

Transcription:

GPhA Fall Technical Conference Nov 2-5, 2015 Bethesda, MD ICH M7 Guidance Overview and Current FDA Perspectives Stephen Miller, Ph.D. CMC-Lead; Office of New Drug Products Office of Pharmaceutical Quality CDER / FDA 1

Presentation Overview 1. Scope/Content of the ICH M7 Guideline 2. Implementation Periods 3. Current FDA Perspectives 2

General Principles Focus on DNA-reactive impurities, i.e. mutagenic impurities typically positive in the bacterial mutagenicity assay Threshold of Toxicological Concern (TTC) concept applies 1x10-5 lifetime risk Less than Lifetime (LTL) principle applies Clinical development and marketed products with shorter treatment durations have higher acceptable levels Evaluate actual impurities plus risk-based subset of potential impurities 3

Guideline General Framework Sections 1-4 Scope etc. Section 5: Impurity Assessment Section 6: Hazard Assessment Scope, General Principles Considerations for Marketed Products What impurities need to be assessed? actual, potential, degradation products Is the impurity mutagenic? QSAR + Ames Section 9: Documentation Section 8: Control Section 7: Risk Characterization Expectations for regulatory filings Expectations, options for impurity control, lifecycle What is the acceptable intake? (TTC, compound specific, less than lifetime exposures) 4

Scope: Intended New drug substances and new drug products in clinical development and subsequent application for marketing Certain post approval submissions of marketed products and to new marketing applications (drug substance previously approved): Changes to the drug substance Changes to the drug product Changes in clinical use See M7 Appendix 1: Scope Scenarios 5

Considerations For Marketed Products When a new drug substance supplier is proposed, evidence that the drug substance produced by this supplier using the same route of synthesis as an existing drug product marketed in the assessor s region is considered to be sufficient evidence of acceptable risk/benefit regarding mutagenic impurities and an assessment per this guideline is not required. 6

Impurity Assessment Section 5: Drug Substance and Drug Product Impurity Assessment Describes an assessment process for actual impurities plus risk-based subset of potential impurities These structures are then evaluated for mutagenic potential 7

Synthetic Impurities and Degradation Products Category (Section) Synthetic Impurities in DS (5.1) From Starting Material (SM) to DS Degradation Products in DS and DP (5.2) Guidance for Assessment Actual impurities in drug substance that are identified (Note: identification would be expected when impurities exceed ICH Q3A identification thresholds) Potential impurities to assess can include SMs, reagents and intermediates. Potential impurities are those reasonably expected to form during the reaction and are a risk based sub set of all potential impurities Assess risk of carryover into DS of identified impurities in SMs and intermediates, and impurities that are reasonably expected by-products in synthesis route However for SMs introduced late in synthesis, where the route of synthesis of SM is known, the final steps of SM synthesis should be assessed Actual degradation products identified in DS and DP under long-term storage conditions (Note: identification would be expected when impurities exceed ICH Q3A/B identification thresholds) Potential degradation products in DS and DP reasonably expected to form under long term storage conditions (e.g., above ICH Q3A/B identification thresholds at accelerated or confirmatory photo-stability studies) 8

Hazard Assessment Known mutagen - evaluate literature and databases Structure of unknown mutagenicity - perform a computational toxicology assessment using (Q)SAR methodologies that predict bacterial mutagenicity Optionally, perform Ames study Mark Powley s and Naomi Kruhlak s talks Additional flexibility during the implementation period to follow pre-m7 approaches to Hazard Assessment 9

Risk Characterization TTC approach can be generally used for compounds predicted to be mutagenic Less-Than-Lifetime concept now includes marketing phase in addition to clinical development phase Tables 2 and 3 in ICH M7 Dave Green s presentation 10

Table 2: Acceptable Daily Intakes for an Individual Impurity (during clinical development and at marketing)* Duration of treatment < 1 month >1-12 months >1-10 years >10 years to lifetime Daily intake [µg/day] 120 20 10 1.5 Intermittent dosing can be based on the total number of dosing days Compound-specific Acceptable Intake (AI) can be adjusted proportionally for shorter durations 11

The treatment duration categories with acceptable intakes in Table 2 for marketed products are intended to be applied to anticipated exposure durations for the great majority of patients. (Section 7.3.2) Examples of Clinical Use Scenarios (Table on Page 19) 12

Control Options (8.1) Option 1: Monitor the impurity in the drug substance Acceptance criterion at or below the TTC Option 2: Monitor the impurity in intermediate, starting material or in-process control Acceptance criterion at or below the TTC Option 3: Monitor the impurity in intermediate, starting material or in-process control Acceptance criterion above the TTC, with demonstrated understanding of fate and purge and associated process controls Option 4: Design robust process controls to reduce the risk of impurity level above the TTC to negligible 13

Documentation (M7 Section 9) Section 9.1 outlines a risk-based approach to documentation during clinical development phases Section 9.2 outlines recommendations for the marketing application 14

Implementation of M7 Guideline The final (Step 4) version of M7 was published on the ICH website in July 2014. Because of the complexity of the guideline, implementation of M7 is not expected until 18 months after ICH publication (i.e., January 2016). Applicants may adopt all or portions of the M7 guideline at any time (e.g., less-than-lifetime limits, approaches to control, class-specific limits), until January 2016 when full implementation is expected. The following slides show exceptions to 18 month timeline 15

Implementation (cont.) Ames tests should be conducted according to M7 irrespective of the stage of development of the Application. However, Ames tests conducted prior to publication of M7 need not be repeated. When development programs have started phase 2B/3 clinical trials BEFORE publication of M7, these programs can be completed up to and including marketing application submission and approval without following M7 (i.e., M7 does not apply; may follow pre-m7 guidance). When development programs have started phase 2B/3 clinical trials AFTER publication of M7, these programs have the option to implement M7 or choose to follow the 18 month grace period (full implementation in January 2016). 16

Implementation (cont.) Given the complexity and lead-time for development of a commercial manufacturing process, application of M7 to new marketing applications that do not include Phase 2B/3 clinical trials is not expected until 36 months after ICH publication of M7 (e.g., new dosage forms, or new DMFs supporting generic drug applications, may follow pre-m7 guidance until July 2017). The 36 month implementation period is also appropriate for applicable post-approval changes (i.e., may follow pre-m7 guidance until July 2017). 17

Clinical Development (e.g., IND) M7 Implementation Timeline New Marketing Applications Requiring Clinical Efficacy & Safety Data (e.g., NDA) New Marketing Applications Without Clinical E & S Data (e.g., new dosage forms, generic drugs)* Applicable Post-Approval Changes (e.g., new synthetic route) Full implementation of M7 not expected until January 2016 Full implementation of M7 not expected until July 2017 * This will be addressed by regional regulatory processes Programs in Phase 2b or 3 before July 2014 may continue to follow Pre-M7 guidance until the marketing application is submitted and approved 18

FDA Case Studies Presented to show diversity of approaches consistent with M7 guidance Outcomes were specific to these cases, and may not apply to other situations with different mutagenic impurities, risk-benefit to patients, etc The control strategy and its risk-based justification must fit its context for each application or master file 19

Case Study 1 Starting Material Step 1 A Step 2 B Step 3 C Step 4 Final Drug Substance Crude Drug Substance Step 5 [ D ] xtal = crystallization 20

Case Study 1 - continued Starting Material Step 1 A Step 2 B* 1 -sulfonate Step 3 C 1 -chloride Imp * Step 4 Final Drug Substance Crude Drug Substance Step 5 [ D ] 21

Case Study 1 - continued Starting Material Precursors to SM Ar-NO 2 * Ar -NO 2 * Step 1 A Step 2 B* 1 -sulfonate Step 3 1 -chloride Imp * C Step 4 Final Drug Substance Crude Drug Substance Step 5 [ D ] 22

Case Study 1 - continued Starting Material Precursors to SM Ar-NO 2 * Ar -NO 2 * Step 1 A Step 2 B* 1 -sulfonate Step 3 1 -chloride Imp * C Step 4 Final Drug Substance Crude Drug Substance Step 5 [ D ] Spike & Purge studies on nitro and chloride impurities support Option 3 control (at intermediate C) Option 1 control on residual sulfonate intermediate in DS Spike & Purge studies could support Option 3 testing or Periodic Verification Testing with Option 1 23

Case Study 2 SM-1 SM-2 Step 1 A Step 2 precip B Reagent Final Drug Substance SM-3 Step 1 C Step 2 precip D 24

Case Study 2 - continued SM-1 SM-2 Step 1 2 Precursors* to SM-2 A Step 2 precip B Reagent* Final Drug Substance SM-3 Step 1 C Step 2 precip D CH 3 Cl By-product* 25

Case Study 2 - continued SM-1 SM-2 Step 1 2 Precursors* to SM-2 A Step 2 precip B Reagent* Final Drug Substance SM-3 Step 1 C Step 2 precip D CH 3 Cl By-product* Acceptable intake > 1.5 ug/day based on clinical use Structures in 2 steps prior to SMs evaluated for mutagenicity Spike & Purge studies, predicted purge factors, and analytical data support Option 4 control for precursors to SM-2 Spike & Purge plus analysis of CH 3 Cl at D supports Option 4 8 of 8 commercial-scale batches shows Reagent < 10% of AI in drug substance; Spike & Purge; supports Option 4 26

Conclusions FDA Perspectives 1) In NDAs, ANDAs and master files, we are seeing many of the approaches to controlling mutagenic impurities that are consistent with the intent of the M7 guidance Options 1-4 control approaches Less-Than-Lifetime acceptable intakes Compound-specific Permitted Daily Exposures Class-specific acceptable intakes (e.g., mono-chloro alkanes) Intermittent use scenarios 27

Conclusions FDA Perspectives 2) Some types of mutagenic impurities (e.g., nitroaromatics, unsaturated ketones) can in some cases persist through a surprising number of reactions and purge points Value of knowledge for specific impurity/synthesis 3) For possible discussion in Q&A: How have applicants/holders been handling situations where there is a significant difference between the loading dose and the subsequent maintenance doses? 28

29