for Human Pharmaceuticals Kyung-Chul Choi D.V.M., Ph.D.

Slides:



Advertisements
Similar presentations
Pre-clinical and Clinical Regulatory Overview
Advertisements

EPAA Annual conference November Regulatory acceptance of alternative approaches for pharmaceuticals Jean-Marc Vidal Safety & Efficacy of Human Medicines.
Marc Bailie DVM, PhD Director In Vivo Facility Michigan State University, Chief Development Officer Integrated Nonclinical Development Solutions (INDS)
Regulatory Framework Leigh Shaw, Director.
Evaluation of a potential mutagenic MOA based on analysis of the weight of evidence and using the modified Hill criteria Martha M. Moore, Ph.D. Director,
Safety and Extrapolation Steven Hirschfeld, MD PhD Office of Cellular, Tissue and Gene Therapy Center for Biologics Evaluation and Research FDA.
Regulatory Toxicology James Swenberg, D.V.M., Ph.D.
1 Pharmacology/Toxicology information to submit an IND for an anticancer drug.
Mitochondrial Manipulation Technologies: Preclinical Considerations
1 PK/PD modeling within regulatory submissions Is it used? Can it be used and if yes, where? Views from industry 24 September 2008.
U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES NATIONAL INSTITUTES OF HEALTH Working with FDA: Biological Products and Clinical Development Critical Path.
What Do Toxicologists Do?
Stefan Franzén Introduction to clinical trials.
+ Drug Development and Review Process. + Objectives Learn the processes involved in drug discovery and development Define the phases involved in FDA drug.
Anne M. Pilaro, Ph.D. Center for Drug Evaluation and Research FDA
Neonatal/Juvenile Animal Safety Studies Kenneth L. Hastings, Dr.P.H., D.A.B.T. Office of New Drugs, CDER.
Effectiveness Evaluation for Therapeutic Drugs for Non-Food Animals
Food and Drug Administration Preclinical safety data for “first in human” (FIH) clinical trials in healthy volunteer subjects Oncology Drug Advisory Committee.
Training Workshop on Pharmaceutical Development with a Focus on Paediatric Medicines / October |1 | Regulatory Requirement on Dossier of Medicinal.
CDER IND/NDA Reviews Guidance, The Common Technical Document and Good Review Practice John K. Leighton, Ph.D., DABT CDER/FDA.
REGISTRATION OF MEDICINES & PROGRESS WITH RESTRUCTURING THE MCC 1.
Guidance for Industry M4S: The CTD-Safety
Committee on Carcinogenicity (COC) Approach to Risk Assessment of Genotoxic Carcinogens David H. Phillips* COC Chairman Descriptive vs. Quantitative.
29/11/2007KLE University, Belgaum CURRENT PHARMACOLOGY & TOXICOLOGY GUIDLINES FOR PHARMACEUTICAL INDUSTRY Dr. Basavaraj K. Nanjwade M.Pharm., Ph.D.
1 Safety Pharmacology for Oncology Pharmaceuticals at CDER John K. Leighton Associate Director for Pharmacology CDER/OND/OODP.
Stefan Franzén Introduction to clinical trials.
The R&D process Clinical development Andy Gray Consultant pharmacist.
Essential Considerations for Your IND Submission: Objectives and Pitfall Avoidance in Your Preclinical Program Darren Warren.
U.S. Food and Drug Administration Notice: Archived Document The content in this document is provided on the FDA’s website for reference purposes only.
Exploratory IND Studies
Adding Safety Pharm Endopoints To General Tox Studies - II Michael J Engwall, DVM, PhD Principal Scientist Safety and Exploratory Pharmacology Toxicology.
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
CHEE DRUG PRODUCT DEVELOPMENT u Drug ä agent intended for use in the diagnosis, mitigation, treatment, cure, or prevention of disease in man or animals.
DETERMINATION OF RELATED SUBSTANCES & IMPURITIES IN DRUGS BY ICH GUIDE LINES BY M.PAUL RICHARDS.
Preclinical Guidelines: Development of Radioprotective/Mitigative Agents Departments of Dermatology & Radiation Oncology University of Rochester Medical.
MAIN TOXICITY TESTING. TESTING STRATEGIES A number of different types of data are used in order to establish the safety of chemical substances for use.
Prof. Dr. Wolfgang Dekant Department of Toxicology University of Würzburg Germany Risk, Hazard, and Innovation.
The New Drug Development Process (www. fda. gov/cder/handbook/develop
Clinical Pharmacology Subcommittee of the Advisory Committee for Pharmaceutical Science Meeting April Quantitative risk analysis using exposure-response.
Safety Pharmacology Society Webinar Series: Safety Pharmacology Endpoints: Integration into Toxicology Studies Integrating functional CNS observations.
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
Consider Incorporating Respiratory Safety Pharmacology Measurements into Your Next Repeat Dose Toxicology Study September 14, 2012 Jeff Tepper, PhD, DABT.
European Patients’ Academy on Therapeutic Innovation Non-clinical development.
European Patients’ Academy on Therapeutic Innovation The key principles of pharmacology.
Pediatric Subcommittee of the AIDAC October 29-30, Topical Immunosuppressants (Calcineurin Inhibitors) - Animal Toxicology October 30, 2003 Barbara.
Evaluating the Safety of Antimicrobial New Animal Drugs with Regard to Their Microbiological Effects on Bacteria of Human Health Concern Qualitative Antimicrobial.
Clinical Trials - PHASE II. Introduction  Important part of drug discovery process  Why important??  Therapeutic exploratory trial  First time in.
1 Biopharmaceutics Dr Mohammad Issa Saleh. 2 Biopharmaceutics Biopharmaceutics is the science that examines this interrelationship of the physicochemical.
Role of VICH and VICH guidelines in the approval process for veterinary medicinal products David Mackay, European Medicines Agency VICH Workshop – Dar.
Summation of Toxicity Data in Vitic Andrew Thresher
Section I General principle of Pharmacology. Where can you get information about general principle of Pharmacology?  Text books:  Katzung, Basic and.
The process of drug development. Drug development 0,8 – 1 mld. USD.
TOPRA Annual Veterinary Symposium Introduction to the Case Study A presentation by Lesley Reeve, Module leader.
The Stages of a Clinical Trial
Drug Discovery &Development
Regulatory– Terms & Definitions רגולציה - מונחים והגדרות
Nonclinical Information in the Common Technical Document: Opportunities for Content Reuse Peggy Zorn, MPI Research Susan Mattano, Pfizer, Inc.
Regulatory Perspective
Biopharmaceutics Dr Mohammad Issa Saleh.
Safety Tests in Cosmetics
Safety Tests in Cosmetics
CTD Module 4: Non-Clinical Studies SPC Relevant Scientific information
Fundamentals of Electronic Submissions and eCTD
Introduction to Research Methods in Psychology
GL18 (R) – Impurities: Residual Solvents in New Veterinary Medicinal Products, Active Substance and Excipients.
Quality guidelines on impurities
Quality guidelines on impurities
Positive Genetox Findings on a Candidate Pharmaceutical…. Now What
VICH GL 54, Studies to evaluate the safety of residues of veterinary drugs in human food: General approach to establish an Acute Reference Dose (ARfD)
Presentation transcript:

for Human Pharmaceuticals Kyung-Chul Choi D.V.M., Ph.D. ICH Safety Guidelines for Human Pharmaceuticals 2008년 10월 15일 Kyung-Chul Choi D.V.M., Ph.D. College of Veterinary Medicine Chungbuk National University Email: kchoi@cbu.ac.kr Blog: blog.daum.net/vetimmune

ICH Introduction * Source : http://www.ich.org

ICH Introduction * Source : http://www.ich.org

ICH Introduction * Source : http://www.ich.org

ICH Introduction * Source : http://www.ich.org

ICH Guidelines The ICH topics are divided into four categories and ICH topic codes are assigned according to these categories. Quality Guidelines Harmonisation achievements in the Quality area including stability studies, impurities testing and a more flexible approach to pharmaceutical quality based on GMP risk management Safety Guidelines Safety Guidelines to uncover potential risks like carcinogenicity, genotoxicity and reprotoxicity. Recent work : non-clinical testing strategy for assessing the QT interval prolongation liability Efficacy Guidelines The work carried out by ICH under the Efficacy heading is concerned with the design, conduct, safety and reporting of clinical trials. Multidisciplinary Guidelines Cross-cutting topics which do not fit uniquely into one of the Q, S and E categories. It includes MedDRA, CTD and the development of Electronic Standards for the Transfer of Regulatory Information (ESTRI). * Source : http://www.ich.org

ICH Guideline = ICH Products * Source : http://www.ich.org

ICH (http://www.ich.org)

Carcinogenecity Studies ICH Safety Guidelines Carcinogenecity Studies S1A – S1C

Carcinogenecity Studies (S1A) TABLE OF CONTENTS 1. Introduction 2. Historical Background 3. Objective of the guideline 4. Factors to consider for carcinogenicity testing 4.1 Duration and Exposure 4.2 Cause for Concern 4.3 Genotoxicity 4.4 Indication and Patient Population 4.5 Route of Exposure 4.6 Extent of Systemic Exposure 4.7 Endogenous Peptides and Protein Substances or Their Analogs 5. Need for Additional Testing

Carcinogenecity Studies (S1B) TABLE OF CONTENTS 1. OBJECTIVE 2. BACKGROUND 3. SCOPE OF THE GUIDELINE 4. THE GUIDELINE 4.1 Preamble. 4.2 Experimental approaches to testing for carcinogenic potential. 5. MECHANISTIC STUDIES 5.1 Cellular changes. 5.2. Biochemical measurements. 5.3. Considerations for additional genotoxicity testing 5.4. Modified protocols. 6. GENERAL CONSIDERATIONS IN THE CHOICE OF AN APPROPRIATE SPECIES FOR LONG TERM CARCINOGENICITY TESTING 6.1. Information from surveys on pharmaceuticals. 6.2. Potential to study mechanisms. 6.3. Metabolic disposition. 6.4. Practicality. 6.5. Testing in more than one species. 6.6. Exceptions.

Carcinogenecity Studies (S1C) TABLE OF CONTENTS 1. INTRODUCTION 1.1 General Considerations for the Conduct of Dose-Ranging Studies 1.2 Toxicity Endpoints in High Dose Selection 1.3 Pharmacokinetic Endpoints in High Dose Selection 1.4 Criteria for Comparisons of AUC in Animals and Man for use in High Dose Selection 1.5 Saturation of Absorption in High Dose Selection 1.6 Pharmacodynamic Endpoints in High Dose Selection 1.7 Maximum Feasible Dose 1.8 Limit Dose 1.9 Additional Endpoints in High Dose Selection 1.10 Selection of Middle and Low Doses in Carcinogenicity Studies 2. SUMMARY

ICH Safety Guidelines Genotoxicity Studies S2

Genotoxicity Studies (S2) TABLE OF CONTENTS 1. INTRODUCTION 1.1 OBJECTIVES OF THE GUIDELINE 1.2. Background 1.3. Scope of the Guideline 1.4 General Principles 2 THE STANDARD TEST BATTERY FOR GENOTOXICITY 2.1 Rationale 2.2 Description of the Two Options for the Standard Battery 2.3 Modifications to the Test Battery 2.4 Detection of Germ Cell Mutagens 3. RECOMMENDATIONS FOR IN VITRO TESTS 3.1 Test Repetition and Interpretation 3.2 Recommended Protocol for the Bacterial Mutation Assay 3.3 Recommended Protocols for the Mammalian Cell Assays

Genotoxicity Studies (S2) TABLE OF CONTENTS (Cont….) 4. RECOMMENDATIONS FOR IN VIVO TESTS 4.1 Tests for the Detection of Chromosome Damage In Vivo 4.2 Other In Vivo Genotoxicity Tests 4.3 Dose Selection for In Vivo Assays 4.4 Demonstration of Target Tissue Exposure for Negative In Vivo Test Results 4.5 Sampling Times for In Vivo Assays 4.6 Number of Animals Analyzed 4.7 Use of Male/Female Rodents in In Vivo Genotoxicity Tests 4.8 Route of Administration 4.9 Use of Positive Controls for In Vivo Studies 5. GUIDANCE ON EVALUATION OF TEST RESULTS AND ON FOLLOW-UP TEST STRATEGIES 5.1 Assessment of Biological Relevance 5.2 Evaluation of Results Obtained in In Vitro Tests 5.3 Evaluation of Results Obtained from In Vivo Tests 5.4 Follow-up Strategies for Positive Results 5.5 Follow-up Genotoxicity Testing in Relation to Tumor Findings in a Carcinogenicity Bioassay

Toxicokinitics and Pharmacokinetics ICH Safety Guidelines Toxicokinitics and Pharmacokinetics S3A – S3B

Toxicokinitics and Pharmacokinetics (S3A) TABLE OF CONTENTS 1. Introduction 2. The objectives of toxicokinetics and the parameters 3. General principles to be considered 3.1 Introduction 3.2 Quantification of exposure 3.3 Justification of time points for sampling 3.4 Contribution to the setting of dose levels in order to produce adequate exposure 3.5 Extent of exposure assessment in toxicity studies 3.6 Complicating factors in exposure interpretation 3.7 Route of administration 3.8 Determination of metabolites 3.9 Statistical evaluation of data 3.10 Analytical methods 3.11 Reporting

Toxicokinitics and Pharmacokinetics (S3A) TABLE OF CONTENTS (Cont…) 4. Toxicokinetics in the various areas of toxicity testing - specific aspects 4.1 Introduction 4.2 Single-dose toxicity studies 4.3 Repeated-dose toxicity studies 4.4 Genotoxicity studies 4.5 Carcinogenicity (oncogenicity) studies1 4.6 Reproductive toxicity studies 5. Supplementary Notes 6. References (other ICH Guidance)

2. Circumstances Under Which Repeated Dose Tissue Toxicokinitics and Pharmacokinetics (S3B) TABLE OF CONTENTS 1. Introduction 2. Circumstances Under Which Repeated Dose Tissue Distribution Studies Should be Considered 3. Design and Conduct of Repeated Dose Tissue Distribution Studies 4. Summary

ICH Safety Guidelines Toxicity Testing S4

TABLE OF CONTENTS (Cont…) 1. Objectives 2. Scope 3. Background Toxicity Testing (S4) TABLE OF CONTENTS (Cont…) 1. Objectives 2. Scope 3. Background 4. GUIDANCE ON DURATION OF CHRONIC TOXICITY TESTING FOR TRIPARTITE DEVELOPMENT PLAN

Reproductive Toxicology ICH Safety Guidelines Reproductive Toxicology S5

Reproductive Toxicology (S5) TABLE OF CONTENTS 1. Introduction 1.1 Purpose of the Guideline 1.2 Aim of studies 1.3 Choice of studies 2. Animal Criteria 2.1 Selection and number of species 2.2 Other test systems 3. General Recommendations Concerning Treatment 3.1 Dosages 3.2 Route and frequency of administration. 3.3 Kinetics 3.4 Control groups 4. PROPOSED STUDY DESIGNS - COMBINATION OF STUDIES 4.1 The most probable option 4.2 Single study design (rodents) 4.3 Two study design (rodents) 5. STATISTICS

Biotechnological Products ICH Safety Guidelines Biotechnological Products S6

Biotechnological Products (S6) TABLE OF CONTENTS 1. INTRODUCTION 1.1 Background 1.2 Objectives 1.3 Scope 2. SPECIFICATION OF THE TEST MATERIAL 3. PRECLINICAL SAFETY TESTING 3.1 General Principles 3.2 Biological Activity/Pharmacodynamics 3.3 Animal Species/Model Selection 3.4 Number/Gender of Animals 3.5 Administration/Dose Selection 3.6 Immunogenicity 4. SPECIFIC CONSIDERATIONS 4.1 Safety Pharmacology 4.2 Exposure Assessment 4.3 Single Dose Toxicity Studies 4.4 Repeated Dose Toxicity Studies 4.5 Immunotoxicity Studies 4.6 Reproductive Performance and Developmental Toxicity Studies 4.7 Genotoxicity Studies 4.8 Carcinogenicity Studies 4.9 Local Tolerance Studies

ICH Safety Guidelines Pharmacology Studies S7A – S7B

Pharmacology Studies (S7A) TABLE OF CONTENTS 1. INTRODUCTION 1.1 Objectives of the Guideline 1.2 Background 1.3 Scope of the Guideline 1.4 General Principle 1.5 Definition of Safety Pharmacology 2. GUIDELINE 2.1 Objectives of Studies 2.2 General Considerations in Selection and Design of Safety Pharmacology Studies 2.3 Test Systems 2.4 Dose Levels or Concentrations of Test Substance 2.5 Duration of Studies 2.6 Studies on Metabolites, Isomers and Finished Products 2.7 Safety Pharmacology Core Battery 2.8 Follow-up and Supplemental Safety Pharmacology Studies……..

Pharmacology Studies (S7B) TABLE OF CONTENTS 1. INTRODUCTION 1.1 Objective of the Guideline 1.2 Background 1.3 Scope of the Guideline 1.4 General Principles 2. GUIDELINE 2.1 Objectives of S7B Studies 2.2 Considerations for Selection and Design of Studies 2.3 Non-clinical Testing Strategy 2.4 Timing of S7B Non-clinical Studies and Integrated Risk Assessment in Relation to Clinical Development 3. TEST SYSTEMS 3.1 Considerations for Test Systems

Immunotoxicology Studies ICH Safety Guidelines Immunotoxicology Studies S8

Immunotoxicology Studies (S8) TABLE OF CONTENTS 1. INTRODUCTION 1.1 Objectives of the Guideline 1.2 Background 1.3 Scope of the Guideline 1.4 Overview 2. GUIDELINE 2.1 Factors to Consider in the Evaluation of Potential Immunotoxicity 2.2 Weight of Evidence Review 3. SELECTION AND DESIGN OF ADDITIONAL IMMUNOTOXICITY STUDIES 3.1 Objectives 3.2 Selection of assays 3.3 Study Design 3.4 Evaluation of Additional Immunotoxicity Studies and Need for Further Studies 4. TIMING OF IMMUNOTOXICITY TESTING IN RELATION TO CLINICAL STUDIES

Nonclinical Evaluation for Anticancer Pharmaceuticals S9 ICH Safety Guidelines Nonclinical Evaluation for Anticancer Pharmaceuticals S9

Nonclinical Evaluation for Anticancer Pharmaceuticals (S9) TABLE OF CONTENTS 1. INTRODUCTION 1.1 Objectives of the Guideline 1.2 Background 1.3 Scope 1.4 General Principles 2. STUDIES TO SUPPORT NONCLINICAL EVALUATION 2.1 Pharmacology 2.2 Safety Pharmacology 2.3 Pharmacokinetics 2.4 General Toxicology 2.5 Reproduction Toxicology 2.6 Genotoxicity 2.7 Carcinogenicity 2.8 Immunotoxicity 2.9 Photosafety testing

Nonclinical Evaluation for Anticancer Pharmaceuticals (S9) TABLE OF CONTENTS (Cont…) 3. NONCLINICAL DATA TO SUPPORT CLINICAL TRIAL DESIGN AND MARKETING 3.1 Start Dose for First Administration in Humans 3.2 Dose Escalation and the Highest Dose in a Clinical Trial 3.3 Duration and Schedule of Toxicology Studies to Support Initial Clinical Trials 3.4 Duration of Toxicology Studies to Support Continued Clinical Development and Marketing 3.5 Combination of Pharmaceuticals 3.6 Nonclinical Studies to Support Trials in Pediatric Populations 4. OTHER CONSIDERATIONS 4.1 Conjugated Products 4.2 Liposomal Products 4.3 Evaluation of Drug Metabolites 4.4 Evaluation of Impurities

Photosafety Evaluation ICH Safety Guidelines Photosafety Evaluation S10

Photosafety Evaluation (S10) TABLE OF CONTENTS 1. INTRODUCTION 1.1. Objectives of the Guideline 1.2. Background 1.3. Scope of the Guideline 1.4. General Principles 2. FACTORS TO CONSIDER IN THE PHOTOSAFETY EVALUATION 2.1. Photochemical Properties 2.2. Tissue Distribution/Pharmacokinetics 2.3. Metabolite Considerations 2.4. Pharmacological Properties 3. NONCLINICAL PHOTOSAFETY TESTING 3.1. General Considerations 3.2. Photoreactivity Testing Using Chemical Assays 3.3. Phototoxicity Testing Using in vitro Assays 3.4. Photosafety Testing Using in vivo Assays and Systemic Administration 3.5. Photosafety Testing Using in vivo Assays and Dermal Administration 3.6. Photosafety Testing Using in vivo Assays and Ocular Administration

Photosafety Evaluation (S10) TABLE OF CONTENTS (Cont…) 4. CLINICAL PHOTOSAFETY ASSESSMENT 5. ASSESSMENT STRATEGIES 5.1. Recommendations for Testing of Pharmaceuticals Given via Systemic Routes 5.2. Recommendations for Testing of Pharmaceuticals Given via Dermal Routes 5.3. Recommendations for Testing of Pharmaceuticals Given via Ocular Routes