Starting Material Selection for Type II Drug Master Files

Similar documents
Regulatory Starting Materials An FDA Perspective

Drug Substance Review in the Office of Pharmaceutical Quality

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

ICH Q11 Questions & Answers Selection & Justification of Starting Materials. Q11 Implementation Working Group 22 May 2018

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

API Selection of Starting Materials Impact on First Cycle Approval

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

Current version 13 October 2016

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

Regulatory Review Considerations of Drug-Linker Quality in ADCs

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

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

CMC Considerations for 505(b)(2) Applications. Monica Cooper, Ph.D. FDA/CDER/OPS/ONDQA AAPS Annual Meeting Washington, D.C.

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

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

2017 AAM CMC Workshop

Commonly Seen Drug Product Related Quality Deficiencies

Evolution of the CMC Review - ANDAs

Guidance for Industry

Future of Question-based Review and Regulatory Submissions

While the recognition

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

EU Regulatory Perspective and Expectations. Sven-Erik Hillver Senior expert, QWP delegate Medical Products Agency, Sweden

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

How we set specifications for impurities (including Genotoxic impurities) 24 May 2017 Elisabeth Kovacs, Apotex CSO Chemistry and Analytical Sci.

Scientific Considerations for Continuous API Manufacturing

Implementation strategy of ICH Q3D guideline

CEP submission: How to prepare a New Application?

Pharmaceutical quality requirements

Guidance for Industry

European reflections on reviewing NDAs and ANDAs for ICH Q3D elemental impurity compliance

Quality of raw materials and manufacturing of advanced therapies. Fouad Atouf, Ph.D. Head, Global Biologics July 12, 2018

A Generic Industry Perspective on Establishing Impurity Limits And a Corresponding Control Strategy

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

The place of the Certification Procedure in the global regulatory environment

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

M7 Assessment and Control of DNA Reactive (Mutagenic) Impurities in Pharmaceuticals to Limit Potential Carcinogenic Risk

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

The Role of Chemists in the FDA Drug Approval Process

Preventing, Detecting, and Responding to Data Integrity Events. Background, Recent Findings, and Agency Expectations

Strategies for IND Filing Success: Chemistry, Manufacturing and Controls

International Pharmaceutical Excipients Council Of The Americas

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

Quality Agreement. by and between. Supplier Name. Address: and. Client Name: Address:

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

FDA Perspective on Standards and Recommendations for Control of Elemental Impurities in Drug Products

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

Implementation strategy of ICH Q3D guideline

Impurities from degradation of Drug Substances

GMPs for Method Validation in Early Development: An Industry Perspective (Part II)

CGMP Requirements for Investigational Products

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

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

Cleaning and Cleaning Validation of API Plant and Equipment

Jeff Yuen and Associates, Inc. PO Box 6026 Orange, CA Zenaida Power Regulatory Expertise:

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

ICH Topic Q 3 A Impurities Testing Guideline: Impurities in New Drug Substances

Introduction to CMC Regulatory Affairs

FDA Guidance and Current Experience with New Drug Submissions

Key-points of Post approval variation in Korea

Pre-Approval Inspection Program Update

Examples of regulatory expectations for analytical characterization and testing

TECHNICAL AND REGULATORY CONSIDERATIONS FOR PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT Q12

Guideline on assessment and control of DNA reactive (mutagenic) impurities in veterinary medicinal products

Toxicological studies of impurities and degradation products: in silico Methodologies as a safety assessment tool

ICH guideline M7 on assessment and control of DNA reactive (mutagenic) impurities in pharmaceuticals to limit potential carcinogenic risk

ICH Quality Implementation Working Group POINTS TO CONSIDER

CANADA (HEALTH CANADA)

Divi's Laboratories Ltd. (Unit II) 4/13/17

Divi's Laboratories Ltd. (Unit II) 4/13/17

Synopsis: FDA Process Validation Guidance

2.3.S.2.5. Process Validation and/or Evaluation

TECHNICAL AND REGULATORY CONSIDERATIONS FOR PHARMACEUTICAL PRODUCT LIFECYCLE MANAGEMENT Q12

Latest Trend of Drug Quality in Korea In-Kyu Kim, Ph.D. Division Director Drug Evaluation Department

US FDA: CMC Issues for INDs

Linking Regulatory Commitments to Post Approval Changes Robert Iser

9/2/2014. USP Monograph Modernization. Todays topics. USP basic. Todays topics. - USP basic. - USP publications. - USP monograph modernization

PAI Inspections, Observations and Data Integrity

Guidance for Industry

POLICY AND PROCEDURES. Office of Pharmaceutical Quality. Table of Contents

Guidance for Industry ANDAs: Stability Testing of Drug Substances and Products

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

An Overview of IQ s Position Paper: Early Development GMPs for Small-Molecule Specifications

2nd FDA/PQRI Conference on Advancing Product Quality

Generics Perspective: Success Strategies for Genotoxic Impurity Identification, Assessment and Control

CATEGORY Advertising. CATEGORY Biopharmaceutics. CATEGORY Biosimilarity

COMMON DEFICIENCIES IN FINISHED PHARMACEUTICAL PRODUCT (FPP) DOSSIERS

Risk based approaches for a sterility assurance application review: a microbiologist s perspective Dr. Yeissa Chabrier Roselló

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

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

Inspections, Compliance, Enforcement, and Criminal Investigations

Impact factor: 3.958/ICV: 4.10 ISSN:

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

Inspection. Implementation of ICH Q8, Q9, Q10

Drug Impurities: The Good, Bad and Ugly

GMP The Other Side of Chemistry, Manufacturing & Controls (CMC)

Q & A on USP General Chapter <823> Revision Proposal Webinar held on 21 February 2011

Regulations of Biologics in Taiwan

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

Transcription:

Starting Material Selection for Type II Drug Master Files Ronald S. Michalak Quality Assessment Lead (Acting), Division of Lifecycle API Office of New Drug Products, OPQ/CDER/ FDA

CDER Reorganization Office of Pharmaceutical Quality In Operation since Jan 11, 2015 Combines Former Office of Pharmaceutical Science (OPS), Office of Compliance and DMF Review Staff from Office of Generic Drugs

The Office of Pharmaceutical Quality Immediate Office PMAS Staff Office of Program and Regulatory Operations Office of Policy for Pharmaceutical Quality Office of Biotechnology Products Office of New Drug Products Office of Lifecycle Drug Products Office of Testing and Research Office of Process and Facilities Office of Surveillance

Office of New Drug Products Office of the Director Division of New Drug API: NDA/IND Drug Substance Review Division of Lifecycle API: DMF Staff/ANDA Drug Substance Review Division of Biopharmaceutics Division of New Drug Products I Division of New Drug Products II

ONDP Drug Substance Review Division of New Drug API 2 Branches each aligned with the OND Clinical Divisions Review of Drug Substance sections of NDAs, INDs and Meeting Packages Division of Lifecycle API 3 Branches Review of DMFs primarily in support of ANDAs (generics)

ONDP Drug Substance Review Reviewers trained in organic/analytical/process chemistry, many staff with industrial experience Drug substance reviews are performed by approximately 80 reviewers (20 for New Drugs and 60 for Lifecycle) One Office improves communication regarding starting material selection between RLD and subsequent lifecycle submissions DMF Scientific Review Process: Primary review Secondary review Tertiary review (if 1 st generic or high risk application) DMF is then adequate, inadequate, or adequate with information request(s)

What is a Regulatory Starting Material? High level definition in ICH Q7 and Q11 Starting point for cgmp manufacturing process Starting point for the detailed description of the synthetic process in a marketing application One or more starting materials is possible depending on linear or convergent syntheses Can be a commercially available (commodity) chemical manufactured mainly for the non-pharmaceutical market Can be synthesized in-house or outsourced Has defined chemical structure

Dilemma: How do we balance starting material selection that impacts industry concern for supply chain flexibility and cost with adequately low risk to quality and patient?

Why do Regulators Request Longer GMP Synthetic Routes? Information on earlier synthetic steps is necessary in order to understand the risk of impurity carryover and to demonstrate that the proposed control strategy sufficiently mitigates this risk A sufficient number of purification steps need to be documented so that the fate and purge of impurities can be understood Recrystallizations and salt formation are not considered chemical transformation steps Multiple synthetic transformations carried out in one vessel without intermediate isolations (sometimes referred to as telescoped steps or one pot reactions ) provide fewer opportunities for purification than if isolation of intermediates were carried out For these scientific reasons, short synthetic routes will not normally be accepted

Regulatory Perspective On considerations for starting material selection In Lifecycle API: (DMF scientific review) Q7 and Q11 principles Commercial availability Information known to the Agency. Is the starting material complex? How is stereochemistry or regiochemistry established Where in the process is the core of the structure formed? Was information provided on how the starting material is made so that the impurity profile can be adequately assessed? What is the justification for the SM specification? Justified limits based on observed impurity values during process validation (retrospective) Spike and purge studies at impurity limits (prospective)

Starting Material Specification Example In-House Specification for Starting Material 1 Test / Method Acceptance Criteria Description White to Off White Solid ID / HPLC RT of main peak matches RS Water / KF NMT 0.50% Purity / HPLC NLT 90.0% Any Single Unspecified NMT 2.0% Impurity/HPLC Total Impurities/HPLC NMT 5.0% No specific ID test Are impurity limits justified Does purity limit reflect observed values in COAs Is test omission for residual solvents justified

Guidance Documents for Selection of ICH Q11 Starting Materials Guidance on Drug Substances with a sub-section on Starting materials ICH Q11, Q & A (in progress) Companion document to Q11 providing additional clarification ICH Q7A Guidance for GMP from the starting material to API

ICH Q11 Overview of Q11 Principles A multi-step GMP process is clearly implied Process risk inherently higher the closer the regulatory starting material is to the final API GMP should be applied to critical process steps which impact the quality of the API Steps that introduce key structural features Steps where careful control of stoichiometry, temperature, other process variables is crucial for API quality Complex chemical transformations

Overview of Current Guidances ICH Q11 Seven General Principles The manufacturing route begins from the starting material and GMPs apply from there onwards; The changes in material attributes and process conditions at the beginning of the manufacturing process have a lower potential to impact API quality; Manufacturing steps that impact the impurity profile of the API should be included in the process description; Enough of the manufacturing process must be disclosed so impurity fate and purge can be understood; Starting materials should be a substance of defined chemical properties, and nonisolated intermediates are not appropriate; Starting materials should be a significant structural fragment and should be distinguished from reagents and solvents; and Commercially available commodity chemicals need not be justified as a starting material as long as these are available in the pre-existing non-pharmaceutical market.

ICH Q11 Clarification of Q11 Principles Starting materials should be a significant structural fragment.. Should not be interpreted to imply close structural similarity between the proposed starting material and the API Intention was to distinguish between raw materials and starting materials Commercially available commodity chemicals need not be justified as starting materials Provide supporting documentation regarding the nonpharmaceutical market for these chemicals Procuring intermediates from vendors is not commercial availability

Clarification of Q11 Principles - Manufacturing steps that impact the impurity profile of the API should be included in the process description - Enough of the manufacturing process must be disclosed so impurity fate and purge can be understood Is enough of the process covered under GMP that we are confident that the process can purge all impurities resulting from the starting material to the specification levels in the final API? Tip: Include a narrative description of the generation, fate, and purge of impurities in the process

Fate & Purge of Process Impurities SM Intermediate I Intermediate II API Impurity A Impurity B Intermediate I Impurity B Impurity C Impurity C Impurity C M7 Impurity M7 Impurity M7 Impurity (<30% TTC) Impurity F Impurity F Impurity F Red > identification threshold Blue < identification threshold

Temporary slide to justify late stage SM s What is a Justified SM in this Route? A + B C D [H] FI DS E F G J DS (salt) Candidate Starting Materials? Salt conversions not considered as chemical transformations(q11) Final Intermediate rarely adequate as SM Unisolated intermediates (H) do not have defined chemical properties J is also a starting material as its structural elements are incorporated into the final API and should be evaluated by Q7 and Q11 principles 18

FDA Concerns Regarding Starting Material Designation Starting material is not manufactured under GMP Change Control and Vendor Qualification Outsourcing is common Cost benefit to industry / High risk for regulators Changes to manufacturing process of starting material and impact on drug substance Risk to impurity control strategy (genotoxic, regioisomers, chirality, heavy metals, etc.) Applicant should have a strong Quality Management System (vendor qualification) (not in scope of review)

Common Issues with Starting Material Designation by Industry Proposing starting materials only one or two steps from the API in a multi-step synthesis Justified by starting material contributing a significant structural fragment ; by this logic even the final intermediate could be a starting material. Contradicts all other Q11 principles and Q7A. Proposing starting materials multiple steps from the API, however: The steps are simple chemical manipulations e.g., esterification, removal of protecting groups. The starting material itself is synthesized by complex reactions, e.g., stereospecific reactions, complex ring systems, etc. The process is telescoped: few or no isolated intermediates

Common Issues with Starting Material Designation by Industry Proposal of an advanced intermediate as a starting material and making a commitment not to change the manufacturing process for it without FDA notification. The advanced intermediate should still meet all criteria for SM outlined in Q11 and Q7A According to interpretation of regulations 21 CFR 314.70 and 21 CFR 314.420 (c), any change could include starting material vendors with a new route of synthesis which results in the addition of tests to the starting material specification. These types of proposals are evaluated on a case-by-case basis

Other Recommendations Justification why steps prior to the starting material, which are not carried out under GMP, are not likely to affect the quality of the API should always be provided. It is strongly recommended that industry seek FDA input on SM selection: For Generics/DMF holders by email at CDER-OPQ-QUERIES@FDA.HHS.GOV

Starting Material Selection: Bottom Line All the general principles in Q11 and Q7A for selection of starting materials should be considered rather than applying any individual one in isolation Commercial availability Complexity of the process and structure Impurity fate and purge Significant structural fragment Adequate SM specification Deficiencies for inadequate SM are common during DMF reviews (both CA and full scientific review) and can be costly: Extra review cycles More work (validation/method development) Risk of RTR for the referencing ANDA