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

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

Starting Material Selection for Type II Drug Master Files

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 Over time, 466 may be used less frequently and may be withdrawn.

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

Flexible and Pending Monographs

Guidance for Industry

Evolution of the CMC Review - ANDAs

Overview of USP Activities and How to Get Involved

Clinically Relevant Dissolution Specifications: FDA Perspective and Initiatives

Regulatory Starting Materials An FDA Perspective

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

IMPURITIES IN NEW DRUG PRODUCTS

PHARMACEUTICAL TESTING

New Drug Product Impurities

Derivation and Justification of Safety Thresholds

Further Stability Considerations

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

CANADA (HEALTH CANADA)

Implementation of the ICH Q3D guideline in the Ph. Eur.

Role of USP Monographs and. General Chapters. Steve Zigler, Ph.D.

API Selection of Starting Materials Impact on First Cycle Approval

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

CDER 2016 Actions and 2017 Priorities. Richard Moscicki Deputy Center Director for Science Operations, CDER, FDA

Reference Standard Characterization. Steve Lane. General Manager, NSF Reference Standards.

ICH Q12 Technical and Regulatory Considerations for Pharmaceutical Product Lifecycle Management

Identifying and Controlling CPPs and CMAs

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

The Role of Chemists in the FDA Drug Approval Process

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

Risk Management: FDA and Industry Experience. Dan Snider, Ph.D Vice President Morgantown RD Mylan Pharmaceuticals Inc.

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

API Stability Protocols and. Chris Byrne Tasmanian Alkaloids

Latest USP Initiatives: Monographs, General Chapters, and Compounding

Drug Substance Review in the Office of Pharmaceutical Quality

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

Early Development Best Practices for Stability- Regulatory Perspective

Analytical Methods Development and Validation

Continuous Manufacturing Achieving the Vision of Modernizing Pharmaceutical Manufacturing

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

Replacing Analytical Methods for Release and Stability Testing CBER Perspective

"NOT FOR IMPLEMENTATION" GUIDANCE FOR INDUSTRY

Guidance for Industry

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

Case study 2: Parenteral Drug Product

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

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

for IND and RDRC Regulated PET Compounding

Future of Pharmaceutical Quality and the Path to Get There

Updates on Quality Metrics - Industry Perspectives Dan Snider Vice President, Research and Development Mylan Inc.

Managing Quality in Pharmaceutical Industry Using Six Sigma. Edited by Mahmoud Farouk Moussa TQM, CSSBB, MBA

Variation Regulations (EU)

Extractables and leachables: An Introduction

Harmonizing FDA Regulation and the Practice of Pharmacy: Challenges and Opportunities. Plus an update on PET Drug User Fees

CDER /OPQ Office of Surveillance Quality Metrics in Surveillance

Elemental Impurities: An Industry Perspective

COMMERCIAL PRODUCT STABILITY

á232ñ ELEMENTAL IMPURITIES LIMITS

22 January Division of Dockets Management (HFA 305) Food and Drug Administration 5630 Fishers Lane, rm Rockville, MD 20852

Laboratory OOS Investigations The Missing Link

CEP submission: How to prepare a New Application?

How to implement ICH Q3D of elemental impurities in 5 steps

Leachable and Extractable Testing

IVT Laboratory Week Jerry Lanese. Ph.D. The Lanese Group, Inc The Lanese Group, Inc.

Current Topics For Sterile Generic Drug Products

QbD implementation in Generic Industry: Overview and Case-Study

STANDARD OPERATING PROCEDURES. Handling and working with Analytical Standards

How to Identify Critical Quality Attributes and Critical Process Parameters

Industry Perspective on Manufacturing in Early Development

Multiple stakeholders; one objective. Atypical Actives Industry Perspective. USP Excipients Stakeholder Forum November 29, 2017

GDUFA RESEARCH AND REGULATORY INITIATIVES FOR COMPLEX TOPICAL PRODUCTS

Regulatory Review Considerations of Drug-Linker Quality in ADCs

Session 7 Clinical Trial Assessment Bioequivalence Studies

CGMP for Phase 1 INDs

USA Proprietary Name Review Process

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

Guideline on setting specifications for related impurities in antibiotics

CLEANING VALIDATION WITH RISK ASSESSMENT

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

Current Features of USFDA and EMA Process Validation Guidance

GUIDELINE FOR THE STABILITY TESTING

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

Inspections, Compliance, Enforcement, and Criminal Investigations

BMS Experience with the FDA-EMA QbD/PAT Joint Pilot Ambarish K. Singh, PhD Director, Global Regulatory Sciences-CMC Bristol-Myers Squibb Company

PROPOSAL FOR REVISION OF THE SUPPLEMENTARY GUIDELINES ON GOOD MANUFACTURING PRACTICES: VALIDATION, APPENDIX 7:NON-STERILE PROCESS VALIDATION

Pre-Approval Inspections for Drug Products

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

Implementation of ICH Q3D in the Certification Procedure

Proposed New USP General Chapter: The Analytical Procedure Lifecycle 1220

Examples of regulatory expectations for analytical characterization and testing

Particulate Matter and Visual Inspection: Industry Trends 2015

Q10 PHARMACEUTICAL QUALITY SYSTEM

Daniel Y. Peng, Ph.D.

Continuous Pharmaceutical Manufacturing: CGMP, Process Validation, and Inspectional Considerations for Implementation

Responding to an FDA 483

Connection Between Quality, Safety, and Efficacy

STIMULI TO THE REVISION PROCESS


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

FDA Process Validation

Transcription:

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

The information within this presentation is based on the presenter's expertise and experience and represents the views of the presenter for the purposes of stimulating discussion at this workshop.

We all aim to do the right thing PAGE 3

We all aim to do the right thing Clinical Relevance Context, Connections and Collaboration FDA/PQRI Conference March 22, 2017 Sarah Pope Miksinski Director (acting), Office of Surveillance Director, Office of New Drug Products PAGE 4

Guidances available to industry 1. ICH Q 3 A&B: Impurities in Drug Substance and Drug Products-Identification and qualification threshold based on MDD or MDI 1. ICH Q3 C Residual Solvents- PDE 2. ICH3D- PDE 3. ICH M7- TTC, duration of treatment, structural similarity to API 4. FDA Guidance for ANDA ( API and Product) +RLD and USP 5. FDA Guidance on Refuse to Receive (RTR) (same as #5) PAGE 5

Clinically Relevant Specifications *Clinically Relevant Specifications ( CRS) Test Methods and acceptance criteria that identify and reject/accept drug product batches that are likely to perform inadequately / adequately in the indicated population (s) The CGMP regulations require that manufacturing processes be designed and controlled to assure that in-process materials and the finished product meet predetermined quality requirements and do so consistently and reliably.** Process controls address variability to assure quality of the product. Controls can consist of material analysis and equipment monitoring at significant processing points ( 211.110(c)). ** Predetermined Quality Requirement = Clinically Relevant Specifications *Richard (Rick) Lostritto, Ph.D CDER/FDA, PQRI, Conference on Evolving Product Quality, September 16-17, 2014, Bethesda, Maryland ** Guidance for Industry Process Validation: General Principles and Practices U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER) Center for Veterinary Medicine (CVM) January 2011 Current Good Manufacturing Practices (CGMP) Revision 1 PAGE 6

ICH Guidelines Clinically relevant Specifications RLD Qualification ( impurities exceeding qualification threshold) Establishing Limits for impurities USP Qualification of Identified impurities Limits unidentified impurities to levels below the ICH level No Reporting Threshold Batch Data ( process capability) 5/23/2017 Progressing through the process limits tend to become increasingly tighter 7

Not Negotiable Patient Risk I bit of reality check Fact At the time Of filing Risk to Quality #1 #2 #3 #4 #5 #6 #7 #8 Limited no of batches (pilot) Limited stability data ( 6 Mo) Limited no of API batches No experience with scale up No experience with commercial process No information about RLD at expiry or the variability of the RLD API may be using a different synthetic route Product manufacturing process may be different than the RLD leading to different impurities ( degradation products) 5/23/2017 8

If the quality of a product confirmed by meeting the specification, than a lot depends on how we establish specifications Acceptance criteria based on process capability Acceptance criteria based on safety (Clinically Relevant Specifications) Potentially reject acceptable batches (failed batches, stability failures, recall, reduced shelf life) Specifications design based on product/ process understanding following scientific risk based principles linked to patient safety Impact on process capability (introduce variation at a level that has is not linked to quality) no value added Impact on process capability ( controls variability at levels relevant for product quality) Specifications can be too wide, to tight, or altogether irrelevant Cannot and do not improve the quality of a drug substance or product by tightening a specification This can only be done by changing the production process 5/23/2017 9

CRS vs Specifications established based on Process Capability Good Product, Good Process CRL UL: 0.5% Bad Process Bad Product? Batch data UL: 0.3% Batches 1->n PAGE 10

Considerations for RLD Limited number of batches available ( variability?) Typically at or around filing the RLD has years before expiring Do the levels observed in the RLD represent safety or process capability? Qualification? Metabolites Chiral Impurities PAGE 11

Lot# Other opportunities: Setting Specifications Solvent: methanol ICH Limit ( ICH Q3C): Option 1 (based on 10g/day intake) 3000 ppm Option 2: PDE (permitted daily exposure) 30mg/day Drug API: example Maximum daily intake in the example is: 7g/day Results ppm 1 2500 2 2700 3 2750 4 2900 5 2600 6 2100 7 2550 8 2950 9 2900 10 2450 (Method Precision:6 samples) RSD: 6.6% SD= 198 ppm Calibration (6 injections): RSD 3.5% (SD=105ppm) Mean: 2640ppm SD: 259 ppm RSD: 10.9% Considerations for specifications: ICH option 1 : 3000 ppm ICH Option 2 (based on PDE): 4285 ppm PDE 30mg methanol : the maximum methanol content to ensure NMT 30mg daily exposure is 4285 ppm, based on the MDD ( maximum daily dose of example) of 7g/day. In other words, methanol content of NMT 4285ppm will correspond to NMT 30mg of methanol daily intake. Options for acceptance criteria: a) 3000 ppm* b) 0.3% c) 0.30% d) 4285 ppm (PDE) e) 0.4% f) 0.40% Acceptance Criteria Probability of being within spec % > USL NMT 3000 ppm 91.76% 8.24% (>8/100) NMT 0.30% 94.33% 5.67% (6/100) NMT 0.3% 99.95% 0.05% (1/10000) NMT 4285 ppm >99.99% 0.00% NMT 0.40% >99.99% 0.00% NMT 0.4% >99.99% 0.00% NA; (>PDE) Points to consider: An acceptance criteria of 3000 ppm will render a result of 3001 OOS An acceptance criteria of 0.3% will be considered in spec up to 0.3499% An acceptance criteria of 0.30 will be considered in spec up to 0.3049% An acceptance criteria 4285 ppm (PDE) will render a result of 4286 ppm as OOS An acceptance criteria of 0.4% will be considered in spec up to 4499 (>PDE) An acceptance criteria of 0.40% will be considered in spec up to 0.4049% Using this mean and SD for a sample of size 1,000,000 random draws from a normal distribution the following can be predicted for future batches: 5/23/2017 12

Generic Product Synthetic route for API is often different from the RLD (at times unique) Different impurities Different form Different solvents Manufacturing Process for the Drug Product Different composition Different manufacturing process PAGE 13

Control Strategy Control Strategy - A planned set of controls, derived from current product and process understanding, that assures process performance and product quality. The controls can include parameters and attributes related to drug substance and drug product materials and components, facility and equipment operating conditions, in-process controls, finished product specifications, and the associated methods and frequency of monitoring and control. (ICH Q10) PAGE 14

Control Strategy for the Generic Product As a result the specifications for impurities can be quite different from the RLD It will require an analytical procedure designed and developed to control impurities that may not be present (or possible) in the RLD Some of the impurities in the RLD may not be present (or possible) in the generic product The USP method may not be suitable, will not detect, will overestimate or underestimated the new impurities ( equivalence?) The Drug Product while it meets ICH may not meet the USP limits using the USP analytical procedure PAGE 15

Generic Product and USP The USP specification for impurities ( analytical procedure and acceptance criteria) are representative of the monograph s sponsor s approved specifications. Are relevant to sponsor's Process ( API and Drug Product) Represent the sponsor s Process Capability The question that needs to be asked: 1. How USP specifications relate to Clinically Relevant Specifications? 2. How accurate are the following: A. USP limits always represent safety limits? B. USP specifications for impurities are always relevant for a generic product? C. The analytical procedure for impurities published in the USP is always appropriate for a generic product? PAGE 16

ICH Reporting Threshold- levels found below reporting threshold are not included in the calculation of total In Drug Product: Synthetic impurities not reported or included in the calculation of total impurities Impurities exceeding identification threshold can be identified and acceptance criteria established accordingly Matrix ( placebo Peaks) are excluded PAGE 17 Other Challenges in establishing specifications USP vs ICH USP No Reporting Threshold (occasional discard limit that only reflect method capability) All detected peaks are reported and included in total impurities No provision to identify impurities found to exceed identification threshold No provision to exclude matrix (placebo peaks)

Paths and processes developed by USP In cases where a product or an API may not meet the USP (current at that time) specifications (with the USP published procedure), USP developed procedures to allow compliance All paths however require FDA approval before it can be published and progress to official text Pending monographs Flexible monographs Interim Revision Procedure USP GC <476> and <1082> Organic Impurities in Drug Substances and Drug Products. 5/23/2017 18

Pending Monographs Three main stages for Pending Monographs: Draft (on USP website; 90-day comment) Authorized (by USP Expert Committee) Official in USP/NF Following publication in PF, the proposals remain in unofficial status until FDA s approval of the market application USP Guideline for Submitting Requests for Revision to USP NF General Information for All Submissions G1.05-00 Page 6 of 7 EFFECTIVE DATE 04/29/2016 by the sponsor. PAGE 19

Flexible Monographs (cont.) General Notices 4.10.10: Applicability of Test Procedures multiple procedures may be included in particular monographs specifically for the purpose of assuring the availability of an appropriate procedure for a particular product. In such cases, a labeling statement to indicate the appropriate application of the procedure(s) will be included in the monograph. A labeling statement is not required if Test 1 is used. PAGE 20

Interim Revision Announcements (IRAs) Interim Revision Announcements (IRAs) are published in Pharmacopeial Forum (PF) as proposed IRAs. After a 90-day notice and comment period and approval by the relevant USP Expert Committee, IRAs are posted as official text on the USP website, and are incorporated into the next available official publication (USP NF or Supplement). PAGE 21

USP GC<476> & <1086) Follows ICH Concepts Identification threshold Qualification threshold Limits based on MDD PAGE 22

RTR Guidance Pursuant to the enactment of the Generic Drug User Fee Amendments of 2012 (GDUFA),7 the Office of Generic Drugs (OGD) is tasked with a number of activities, including the development of enhanced refusal to receive standards for ANDAs and other related submissions by the end of year 1 of the program. Enhanced RTR standards are important because the practice of submitting an ANDA that is not sufficiently complete to permit a substantive review, which then is repaired via several cycles of applicant resubmission and FDA response, is inherently inefficient and a drain on FDA resources. 5/23/2017 23

Closing Remarks The agency is moving toward Clinically Relevant Specifications and guiding industry to consider risk in the context of clinical performance Encourage industry to use science and risk considerations when establishing specifications that should lead to tests and acceptance criteria based on clinical relevance and not on process capability and/or batch data Give industry opportunities to work in the framework of the USP processes that rely on FDA review and approval before changing and/or progressing to official text. PAGE 24

Closing Remarks (cont.) Understand the challenges industry faces when expected to commit to compendia limits when these do not represent safety therefore are not clinically relevant but rather represent the monograph s sponsor process capability Industry would appreciate a more in-depth evaluation during the technical review stage and more flexibility over the enhanced assessment which occurs at filing. Collaborate with industry to move from what appears to be a one measure fits all approach to a more science and risk based process for establishing limits for impurities in products that reflects the innovation in FDA s regulatory approaches. PAGE 25

A Generic Industry Perspective on Establishing Impurity Limits And a Corresponding Control Strategy An applicant is not expected to tighten the limits based on process capability, provided that the elemental impurities in drug products do not exceed the PDE ( ICH Q3D) An applicant is not expected to tighten limits for impurities based on process capability provided that the impurities in drug products do not exceed the qualified levels Is this enlightened philosophy consistently followed by FDA re: organic impurity limits in ANDAs? Nicholas Cappuccino, PhD Vice-President, Head of Quality and Scientific Affairs, Dr. Reddy s Laboratories, Inc. 2016 GPhA CMC Workshop October 2016, Bethesda Marriott 5/23/2017 26

A Generic Industry Perspective on Establishing Impurity Limits And a Corresponding Control Strategy An applicant is not expected to tighten the limits based on process capability, provided that the elemental impurities in drug products do not exceed the PDE ( ICH Q3D) An applicant is not expected to tighten limits for impurities based on process capability provided that the impurities in drug products do not exceed the qualified levels* ICH Q9: PRINCIPLES OF QUALITY RISK MANAGEMENT Two primary principles of quality risk management are: The evaluation of the risk to quality should be based on scientific knowledge and ultimately link to the protection of the patient; and The level of effort, formality and documentation of the quality risk management process should be commensurate with the level of risk. *Nicholas Cappuccino, PhD Vice-President, Head of Quality and Scientific Affairs, Dr. Reddy s Laboratories, Inc. 2016 GPhA CMC Workshop October 2016, Bethesda Marriott 5/23/2017 27

CRS vs Specifications established based on Process Capability Good Product, Good Process CRL UL: 0.5% Bad Process Bad Product? Batch data UL: 0.3% Batches 1->n PAGE 28

CRS vs Specifications established based on Process Capability Good Product, Good Process CRL UL: 0.5% Batches 1->n PAGE 29

We all aim to do the right thing PAGE 30

Thank you for your attention Any Questions? ekovacs@aptex.com Elisabeth Kovacs, Apotex, 150 Signet Drive Toronto Ontario, Canada, M9L 1T9 PAGE 31