26
INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION Aspects of Quality, Safety and Efficacy Editors ANTHONY C. CARTWRIGHT Medicines Control Agency, Department of Health, London and BRIAN R. MATTHEWS Alcon Laboratories Ltd., Croydon, Surrey ELLIS HORWOOD NEW YORK LONDON TORONTO SYDNEY TOKYO SINGAPORE

INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

INTERNATIONALPHARMACEUTICALPRODUCTREGISTRATION

Aspects of Quality, Safety and Efficacy

EditorsANTHONY C. CARTWRIGHTMedicines Control Agency, Department of Health, London

and BRIAN R. MATTHEWSAlcon Laboratories Ltd., Croydon, Surrey

ELLIS HORWOODNEW YORK LONDON TORONTO SYDNEY TOKYO SINGAPORE

Page 2: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents

Preface and acknowledgements xxx

List of contributors xxxviii

PART I CHEMISTRY, PHARMACY AND MANUFACTURING

1 Pharmaceutical development 3M. J. Morris

1.1 Introduction 31.2 Scientific principles 4

1.2.1 Preformulation studies 41.2.1.1 Solubility 41.2.1.2 pH and pKa 51.2.1.3 Particle size 51.2.1.4 Polymorphism and crystal properties 61.2.1.5 Moisture content 7

1.2.2 Liquid dose forms 71.2.3 Semi-solid dose forms 91.2.4 Solid dose forms 101.2.5 Controlled release preparations 12

1.2.5.1 Solid dose oral preparations 131.2.5.2 Liquid preparations 14

1.2.6 Other dose forms 151.2.6.1 Injections and implants 151.2.6.2 Ocular preparations 161.2.6.3 Inhalation therapy 171.2.6.4 Transdermal systems 18

1.3 Regulatory requirements 191.3.1 European Community requirements 191.3.2 United States requirements 23

Page 3: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

vi Contents

1.3.3 Japanese requirements 251.3.4 Requirements in other territories 26

1.4 New developments and conclusions 261.5 References 29

2 Packaging materials 32D. A. Dean

2.1 Introduction 322.2 Packaging materials and their properties 36

2.2.1 Introduction 362.2.2 Glass in packaging 362.2.3 Metals in packaging 372.2.4 Plastics in packaging 382.2.5 Elastomeric materials (mainly used for closures) 392.2.6 Other contact materials 402.2.7 Summary of packaging materials for primary packs . . . . 40

2.3 Regulatory guidelines 412.4 The environmental concerns 432.5 Checking out information 442.6 Packaging material - evaluation and testing with the product . . . 482.7 Packaging material - evaluation and testing (independent of

product) 522.7.1 Tests and evaluations applicable to any material 522.7.2 Tests for specific materials 542.7.3 Biological tests 552.7.4 Tests for elastomers 552.7.5 Microbiological tests 55

2.8 The evaluation of the role of the pack in the administration of theproduct 56

2.9 The evaluation of closures and closure systems 582.10 Conclusions 592.11 Appendices A2.1-A2.7 612.12 References 75

3 Manufacturing 79I. K. Sykes

3.1 Introduction 793.2 EC requirements for manufacturing information on drug products . . 81

3.2.1 Manufacturing formula 813.2.1.1 Starting materials 813.2.1.2 Batch size 823.2.1.3 Batch homogeneity 823.2.1.4 Continuous manufacture 82

3.2.2 The manufacturing process 83

Page 4: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents vii

3.2.2.1 Production flow chart 833.2.2.2 Process description 833.2.2.3 In-process controls 843.2.2.4 Reworking 843.2.2.5 Sterilisation processes 873.2.2.6 Pharmaceutical assembly 90

3.2.3 Validation of the manufacturing process 903.2.3.1 Types of process validation 91

3.2.4 Validation of the manufacturing equipment 933.2.4.1 Installation qualification (IQ) 933.2.4.2 Operational qualification (OQ) 933.2.4.3 Performance qualification (PQ) 93

3.2.5 Validation of procedures 943.2.6 Documentation of validation 94

3.2.6.1 Validation protocol 943.2.6.2 Validation report 953.2.6.3 Process validation documentation 95

3.2.7 Role of the pharmaceutical Expert 973.2.7.1 Method of manufacture 973.2.7.2 Process validation 973.2.7.3 Site of manufacture 98

3.2.8 Inspection 983.3 FDA requirements for manufacturing information on drug products . 99

3.3.1 Format and content of Chemistry, Manufacturing and Controlsection of an NDA 1003.3.1.1 Drug substance 1003.3.1.2 Drug product 1003.3.1.3 Environmental assessment 102

3.3.2 Summary to an NDA 1033.3.3 Inspections 1043.3.4 Post-licensing data 106

3.4 Biological and biotechnology-derived products in the EC 1073.4.1 Site of manufacture 1093.4.2 Manufacturing process 109

3.4.2.1 Process flow chart 1093.4.2.2 Starting materials 1093.4.2.3 Seed lot system IK)3.4.2.4 Pre-culture 1103.4.2.5 Fermentation I l l3.4.2.6 Harvest 1133.4.2.7 Purification 1133.4.2.8 Primary pharmaceutical operations 1163.4.2.9 Aseptic filling 1173.4.2.10 Lyophilisation 1173.4.2.11 Packaging and labelling 118

Page 5: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

viii Contents

3.4.2.12 Finished product storage 1183.5 The FDA perspective on biological and biotechnology-derived

products 1183.5.1 The Establishment Licence Application 1183.5.2 The Product Licence Application 1243.5.3 Inspection 125

3.6 References 126

4 Active ingredients 129B. R. Matthews

4.1 Introduction: Purpose of this chapter 1294.2 Background 130

4.2.1 What is controlled under the pharmaceutical regulations? . . 1304.2.2 How much information is needed? 131

4.3 Data requirements: 'New drugs' 1334.3.1 Appropriate, available and acceptable pharmacopoeial

specifications 1334.3.2 Suitability of available data for submission 1334.3.3 Submission of all relevant data 1344.3.4 Analytical validation 1344.3.5 Analytical method description 1354.3.6 Reference materials: samples 1354.3.7 Nomenclature of active ingredients 1354.3.8 Inclusion of data: In the submission or in a Drug Master

File? 1364.4 Data to be submitted 137

4.4.1 Description 1374.4.2 Physical and chemical characteristics 1374.4.3 Nomenclature 1374.4.4 Manufacturer 1374.4.5 Manufacturing or synthetic process, purification, and in-

process controls 1374.4.5.1 Synthesis 1374.4.5.2 Purification and final drying 1394.4.5.3 Other stages 1394.4.5.4 In-process and intermediate controls 1404.4.5.5 Process validation data 140

4.4.6 Development chemistry and proof of structure 1414.4.7 Specification for the active ingredient 1414.4.8 Analytical methods 1434.4.9 Analytical validation 1434.4.10 Stability data 1434.4.11 Reference materials (reference standards) 145

4.5 References 145

Page 6: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents ix

4.5.1 United States 1454.5.2 Japan 1464.5.3 European Community 1464.5.4 ICH proposals 147

5 Excipients 148P. K. Wotton, G. Wade and R. C. Moreton

5.1 Introduction 1485.2 Definition and role of excipients 148

5.2.1 Definitions 1485.2.2 Excipients in relation to efficacy 1495.2.3 Safety of excipients 150

5.3 Regulatory and related texts 1515.3.1 International aspects of excipient approval 152

5.3.1.1 United States 1525.3.1.2 Europe 1525.3.1.3 New excipients 1545.3.1.4 Established (non-pharmacopoeial) excipients . . . 155

5.3.2 The ISO 9000-series of quality systems standards 1555.3.3 International aspects: harmonisation 156

5.4 Excipient performance 1585.4.1 Introduction 1585.4.2 Pharmacopoeial tests 1595.4.3 Extra-pharmacopoeial testing 1595.4.4 Performance specifications 1605.4.5 Equivalence of excipient lots from two or more suppliers . . 161

5.4.5.1 Quality 1625.4.5.2 Performance 1625.4.5.3 Dissolution/bioavailability 1635.4.5.4 Stability requirements 1635.4.5.5 Regulatory considerations 1635.4.5.6 Costs 163

5.4.6 Examples 1635.4.6.1 Sodium starch glycollate 1645.4.6.2 Cetostearyl alcohol 1645.4.6.3 Lactose 1645.4.6.4 Magnesium stearate 1655.4.6.5 Hydroxypropyl methylcellulose (hypromellose) . . 165

5.5 What really concerns the regulatory authorities? 1665.5.1 Established, Traditional or Non-critical excipients 1675.5.2 Traditional, Critical excipients 1675.5.3 New excipients 1685.5.4 New, Critical excipients 168

5.6 References 169

Page 7: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

x Contents

6 Control tests on the finished product 172A. Artiges, M.-H. Loulergue and J.-P. Reynier

6.1 Introduction 1726.2 European requirements 174

6.2.1 Harmonisation of national regulations 1746.2.2 Requirements concerning control tests on the finished

product 1756.2.3 How to constitute the dossier 176

6.2.3.1 Administrative environment and regulations . . . 1766.2.3.2 Scientific research and development quality . . . 1796.2.3.3 The transition to production and conformity

quality 1886.3 Japanese and United States (FDA) requirements 191

6.3.1 Specific Japanese requirements 1926.3.1.1 Administrative organisation and general

considerations 1926.3.1.2 Presentation of the data 1926.3.1.3 Relationship with the Japanese Pharmacopoeia

(JP) 1946.3.1.4 Relationship with GMP 1956.3.1.5 Document summary ('Gaiyo') 195

6.3.2 United States requirements - Food and DrugAdministration (FDA) 1966.3.2.1 Administrative organisation and general

considerations 1966.3.2.2 Presentation of the data 1966.3.2.3 Drug Master Files (DMFs) 1996.3.2.4 Relationship with the USP 1996.3.2.5 Verification by the FDA of the methods for control

testing of the finished product 1996.4 Conclusions 2016.5 References 201

6.5.1 European references 2016.5.2 Japanese references 2026.5.3 United States references 203

Appendix A6.1 203European Pharmacopoeia: Dosage forms andtechnological tests 203

Dosage forms (general monographs) 203Dosage form monographs under elaboration 204Technological texts 204Technological texts under elaboration 204

Appendix A6.2 204Japanese Pharmacopoeia: Dosage form general monographs . . . 204

Page 8: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xi

7 Stability data 206A. C. Cartwright

7.1 Introduction 2067.1.1 The purpose of stability testing of medicinal products in

their final packaging 2067.1.2 The purpose of stability testing of active substances . . . . 2077.1.3 The development of stability testing requirements 207

7.2 Scientific and technical background to the design of stability studies . 2097.2.1 Factors influencing the design of stability studies 2097.2.2 Stability testing of products for distribution in the tropics . . 2097.2.3 Classification of climatic zones and choice of testing

conditions 2097.2.4 Kinetics of chemical degradation reactions in pharmaceutical

products 2127.2.5 Chemical stability of solids 2137.2.6 Toxicity of chemical degradation products 2137.2.7 Physical stability 2147.2.8 Influence of packaging on product stability 2157.2.9 Reduced testing designs for stability studies 216

7.2.9.1 Bracketing 2167.2.9.2 Matrixing (fractional factorial design) 217

7.2.10 Stages of development and marketing of products and theneed for stability testing 217

7.3 Regulatory requirements for stability testing in the United States,European Community and Japan 2187.3.1 Testing of New Active Substances (NASs) and other

products - current guidelines 2187.3.1.1 Accompanying data needed when stability data are

submitted 2197.3.2 Testing of bulk active drug substances 220

7.3.2.1 New Active Substances (NASs - substances notpreviously authorised) 220

7.3.2.2 Stability testing of existing (approved) activesubstances 222

7.3.3 Testing of dosage forms in their final packaging 2227.3.3.1 NAS products 2227.3.3.2 Existing active substance products (abridged/

abbreviated products) 2267.3.4 Submission of additional data during the approval process . . 2277.3.5 Stability studies on revised products 22K7.3.6 Routine follow-up testing of production batches of finished

product in its final packaging, and GMP testing 2297.3.7 Extension of shelf life after authorisation of the product . . . 230

Page 9: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xii Contents

7.4 International harmonisation 2307.4.1 New active drug substance testing 232

7.4.1.1 Stress testing of the drug substance 2327.4.1.2 Systematic testing of the drug substance to estimate

the re-test period and recommended storageconditions 232

7.4.2 New active substance product testing 2347.4.3 Other categories of products 237

7.4.3.1 New dosage forms 2377.4.3.2 Generic products 2387.4.3.3 Biological and biotechnological products 239

7.4.4 Light stability testing of products 2397.5 Conclusions 2427.6 References 242

8 Analytical validation 246G. P. R. Carr and J. C. Wahlich (section 8.1)R. J. N. Tanner (section 8.2)8.1 Product and active ingredient related validation 246

8.1.1 Introduction 2468.1.2 Requirements of regulatory authorities 247

8.1.2.1 United Kingdom authority (Medicines ControlAgency) 247

8.1.2.2 United States authority (Food and DrugAdministration) 247

8.1.2.3 European Community (EC) 2478.1.2.4 United States Pharmacopeia 2508.1.2.5 Japanese authority (Ministry of Health and

Welfare, MHW) 2518.1.2.6 Canadian authority (Health Protection Branch,

HPB) 2518.1.2.7 Australian authority (Therapeutic Goods

Authority, TGA) 2528.1.2.8 International Conference on Harmonisation (ICH) . 2528.1.2.9 Conclusions 252

8.1.3 Parameters to be validated for different test methods . . . . 2538.1.3.1 Validation parameters 2538.1.3.2 Selection of parameters for different analytical

methods 2538.1.4 Determinations of validation parameters 254

8.1.4.1 Introduction 2548.1.4.2 Accuracy 255

8.1.4.2.1 Definition of accuracy 2558.1.4.2.2 Determination of accuracy 255

8.1.4.3 Precision 259

Page 10: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xiii

8.1.4.3.1 Definition of precision 2598.1.4.3.2 Determination of precision 259

8.1.4.4 Linearity 2618.1.4.4.1 Definition of linearity 2618.1.4.4.2 Determination of linearity 261

8.1.4.5 Limit of detection (LoD) and Limit of quantitation(LoQ) 2648.1.4.5.1 Definitions of LoD and LoQ 2648.1.4.5.2 Determinations of LoD and LoQ . . . 264

8.1.4.6 Selectivity and specificity 2668.1.4.6.1 Definition of terms 2668.1.4.6.2 Determination of selectivity 2678.1.4.6 3 Determination of specificity 270

8.1.4.7 Analyte and system stability and other phenomena 2708.1.4.7.1 Requirements 2708.1.4.7.2 Potential causes of instability

and other analyte influences 2718.1.4.7.3 Determination and control 271

8.1.4.8 Robustness 2728.1.4.8.1 Definition 2728.1.4.8.2 Determination of robustness 273

8.1.5 Revalidation 2768.1.6 System suitability tests (SSTs) 277

8.1.6.1 Definition 2778.1.6.2 Traditional approaches to SSTs 2798.1.6.3 Individual SSTs 279

8.1.6.3.1 Robustness 2798.1.6.3.2 Accuracy 2798.1.6.3.3 Precision 2808.1.6.3.4 Selectivity 2818.1.6.3.5 Analyte and system stability 2818.1.6.3.6 Linearity 2818.1.6.3.7 Limit of quantitation and

detection 2828.1.6.3.8 Tailing factor 2828.1.6.3.9 Column efficiency 2828.1.6.3.10 Retention time 283

8.1.7 Conclusions 2838.1.8 References 283

12 Pharmacokinetic and toxicokinetic methods validation 2868.2.1 Introduction 286

8.2.1.1 The three phases of the development and applicationof an assay 286

8.2.1.2 Definition of the terms: standard and sample . . . 287

Page 11: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xiv Contents

8.2.1.3 Regulatory requirements 2878.2.2 Assay development 287

8.2.2.1 A structured approach to assay validation . . . . 2878.2.2.2 Selection of a suitable matrix 2878.2.2.3 External and internal standards 287

8.2.3 Assay validation 2888.2.3.1 Documentation of an assay procedure 2888.2.3.2 Calibration standards 2888.2.3.3 Limit of quantification 2908.2.3.4 Accuracy and precision 2918.2.3.5 Linearity 2928.2.3.6 Selectivity 2928.2.3.7 Stability 2948.2.3.8 Recovery 2958.2.3.9 Assay reproducibility 2968.2.3.10 Additional validation for samples in different matrices,

or from different species 2968.2.3.11 Limited re-validation for new staff and of lapsed

assays 2978.2.3.12 Batch size 2978.2.3.13 Additional validation issues for immunoassays . . 2978.2.3.14 The validation report 299

8.2.4 Study monitoring 3008.2.4.1 Introduction 3008.2.4.2 Calibration standards 3008.2.4.3 Quality control samples 3018.2.4.4 Re-analysis of study samples 3028.2.4.5 The bioanalytical appendix 303

8.2.5 Conclusions 3038.2.6 Acknowledgements 3048.2.7 References 304

9 Biopharmaceutics 307

I. J. McGilveray

9.1 Introduction 307

9.2 Biopharmaceutics information to be documented 3109.2.1 Factors affecting dissolution and/or drug absorption . . . . 310

9.2.1.1 Physicochemical characteristics of the active drugsubstance 311

9.2.1.2 Factors related to formulation 3149.2.1.3 Dosage (galenical) form 3169.2.1.4 Manufacturing process 3199.2.1.5 Stability and storage (stability of active substance

and dosage form) 320

Page 12: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xv

9.2.1.6 Dissolution tests and procedures 3209.2.1.7 Regulatory conclusions on physicochemical

factors 3219.2.2 Dissolution standards 3249.2.3 Physiological factors affecting bioavailability 327

9.2.3.1 Permeability and pKa 3289.2.3.2 GI transit/motility 3289.2.3.3 Site-specific absorption or narrow absorption

'window' 3289.2.3.4 Preabsorptive metabolism 3289.2.3.5 Hepatic metabolism 3299.2.3.6 Biliary excretion 3299.2.3.7 Renal excretion 3299.2.3.8 Protein and tissue binding 3299.2.3.9 Regulatory conclusions on physiological factors . . 330

9.3 Pharmacokinetics and bioavailability 3309.3.1 Preclinical aspects of bioavailability 3309.3.2 Clinical pharmacology and bioavailability 3309.3.3 Absolute and comparative bioavailability 3329.3.4 Bioavailability and scale-up from development to

production 3329.4 Bioequivalence 335

9.4.1 Definitions 3359.4.2 Evidence to establish bioequivalence 3369.4.3 Waivers from bioequivalence requirements 3379.4.4 Basis for demonstrating bioequivalence 3389.4.5 Study design, subjects, environment 3399.4.6 Analyte for bioequivalence 340

9.4.6.1 Use of metabolite in bioavailability/bio-equivalence 340

9.4.6.2 Urinary elimination in bioequivalence 3429.4.7 Presentation of bioavailability and bioequivalence data . . . 3429.4.8 Metrics of bioequivalence 3439.4.9 Statistical analysis 345

9.5 Modified release bioavailability/bioequivalence 3479.5.1 Guidance on quality 3489.5.2 Pharmacokinetic and clinical evaluation of modified release

dosage forms 3509.5.3 Food effect investigations with modified release products . . 352

9.6 Conclusions 3549.7 Acknowledgements 3549.8 References 354A.9.1 Appendix 361

Page 13: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xvi Contents

10 Radiopharmaceuticals 364B. R. Matthews

10.1 Introduction 36410.2 Control of radiopharmaceuticals in the European Community . . . 365

10.2.1 Introduction 36510.2.2 The implementation of Directive 89/343/EEC and other

relevant legislation 36610.2.3 Expert Reports 36810.2.4 Application of Article 4 of Directive 65/65/EEC . . . . 36910.2.5 Data requirements 370

10.2.5.1 Introduction 37010.2.5.2 Data requirements 370

10.2.6 Summary of product characteristics 38510.2.7 Labelling requirements 38510.2.8 Instruction leaflet 38610.2.9 Manufacture 387

10.3 Control of radiopharmaceuticals in the United States 38810.3.1 Introduction 38810.3.2 The application for approval for marketing 389

10.3.2.1 The application form 39010.3.2.2 The index 39010.3.2.3 The summary 39010.3.2.4 Technical sections 39010.3.2.5 Environmental impact statement 40210.3.2.6 Labelling 403

10.4 Control of radiopharmaceuticals in Japan 40310.4.1 Introduction 40310.4.2 Data requirements 404

10.4.2.1 General; and chemistry and pharmaceuticaldata 404

10.4.2.2 Preclinical data 40510.4.2.3 Clinical studies 407

10.5 References 407

PART II PRECLINICAL TOXICOLOGY AND PHARMACOLOGY

11 Preclinical testing strategy 411R. Bass and P. Giinzel

11.1 Introduction 41111.2 Preclinical requirements currently in use 41211.3 General co-ordination between preclinical and clinical studies . . 41311.4 General concept of developing preclinical testing strategies . . . 41411.5 General concept of developing individual testing strategies . . . . 415

Page 14: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xvii

11.5.1 Taking into account the main characteristics of envisagedclinical studies 415

11.5.2 Areas of possible human risk to be clarified experimentally 41611.5.3 Mandatory principles and standards 41611.5.4 Technical details of the experiments to be performed . . 416

11.6 The use of flow-charts for the development of preclinical testingstrategies 41611.6.1 The principle of the flow-charts 41811.6.2 Test areas and available flow-charts 420

11.6.2.1 Single-dose toxicity 42311.6.2.2 Toxicity by repeated administration 42311.6.2.3 Toxicity to reproduction 42411.6.2.4 Genotoxic potential 42711.6.2.5 Tumorigenic potential 427

11.7 Conclusions 42811.8 References 428

12 Single-dose and repeat-dose toxicity 430J. C. Topham

12.1 Introduction 43012.2 Single-dose toxicity studies 431

12.2.1 Regulatory considerations 43112.2.2 Outline protocol for a single-dose toxicity study . . . . 43112.2.3 Features of single-dose toxicity study 432

12.2.3.1 Objectives 43212.2.3.2 Species 43312.2.3.3 Sex and number of animals 43312.2.3.4 Route of administration 43412.2.3.5 Dose levels, vehicle and dose volumes . . . . 43412.2.3.6 Observations 43512.2.3.7 Data evaluation and presentation 435

12.2.4 The future 43612.3 Repeat-dose toxicity studies 437

12.3.1 Objectives 43712.3.2 Duration of repeat-dose studies 44012.3.3 Outline protocol for repeat-dose toxicity studies . . . . 443

12.3.3.1 Species 44312.3.3.2 Route of administration 44412.3.3.3 Duration, dose-frequency and drug withdrawal

periods 44512.3.3.4 Number of animals and groups 44512.3.3.5 Dose selection 44612.3.3.6 Pharmacokinetic monitoring and 'toxicokinetics' 44612.3.3.7 Observations 448

Page 15: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xviii Contents

12.3.4 Data evaluation 45012.3.4.1 Study design and statistical considerations . . 45012.3.4.2 Control data 45112.3.4.3 Drug substance 45212.3.4.4 Effect definition 452

12.3.5 Overall assessment of single- and repeat-dose toxicitystudies 454

12.3.6 The future 45512.4 Acknowledgements 45512.5 References 455

13 Reproductive Toxicology 458B. Ulbrich, A. K. Palmer and R. Bass

13.1 Aim of studies 45813.2 Approach to testing 45813.3 Designing studies 460

13.3.1 Selection of species 46013.3.2 Selection of dosages 46113.3.3 Route and frequency of administration 46113.3.4 Kinetics 46213.3.5 Use (and misuse) of preliminary studies 46213.3.6 Group sizes 46713.3.7 Observations 467

13.4 Study options 46713.4.1 Male and female fertility and early embryonic development 468

13.4.1.1 Treatment periods 46913.4.1.2 Mating 469

13.4.2 Effects on pregnancy, conceptus and offspring 46913.4.2.1 Postnatal evaluation 46913.4.2.2 Prenatal evaluation 47113.4.2.3 Special studies (direct treatment of offspring) . . 471

13.5 Data analysis 47113.6 Data presentation 47213.7 Interpretation - extrapolation 47313.8 References 473

14 Mutagenicity 474D. G. Gatehouse

14.1 Introduction 47414.2 Scientific background to field of genotoxicity 475

14.2.1 Mutation 47514.2.2 Mechanisms of mutagenesis 475

14.2.2.1 Events at gene level (point mutations) . . . . 47614.2.2.2 Events at individual chromosome level . . . 476

Page 16: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xix

14.2.2.3 Events at level of the complete genome . . 47714.2.3 Relevance of mutagenicity results to human hazard . 477

14.2.3.1 Germ cell mutations 47714.2.3.2 Somatic cell mutation and cancer 47814.2.3.3 Other possible deleterious effects of somatic

mutations 47914.3 Regulatory requirements 479

14.3.1 Overview 4gO14.3.2 EC requirements 48814.3.3 Japanese requirements 49114.3.4 UK guidelines 49214.4.5 Canadian guidelines 495

14.4 In vitro metabolic activation and the role of in vivo assays . . . . 49714.5 Timing of preclinical genotoxicity tests in relation to Phase I

clinical studies 49914.6 Test systems (protocol variations) 500

14.6.1 In vitro tests for gene mutation in bacteria 50014.6.2 In vitro tests for chromosome damage in mammalian cells . 50814.6.3 In vitro tests for gene mutation in mammalian cells . . . 51514.6.4 In vivo tests for chromosome damage in rodents . . . . 517

14.6.4.1 Micronucleus tests in rodent bone marrow 51714.6.4.2 Metaphase analysis in rodent bone marrow . . 523

14.6.5 Additional in vivo tests for genotoxicity (unscheduled DNAsynthesis assay in rat liver) 524

14.7 Overview of current international initiatives designed to harmonisetest strategies/protocols 52614.7.1 International Conference on Harmonisation (ICH2) . . 527

14.7.1.1 Strategy issues 52814.7.1.2 Test performance 531

14.7.2 International workshop on standardisation of procedures ingenetic toxicology 534

14.8 Present deficiencies and future developments 53714.8.1 Metabolically competent cell lines 53714.8.2 Tests for genome mutations (aneuploidy) 53814.8.3 Transgenic mouse models for use as in vivo somatic

mutation assays 54014.8.4 Single-cell gel electrophoresis assay (comet assay) for the

evaluation of potential organo-specific effects 541

14.9 Conclusions 54114.10 References 542

15 Oncogenicity/carcinogenicity 553B. Rushton

15.1 Introduction 553

Page 17: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xx Contents

15.2 General points on study conduct and interpretation 55415.3 When are carcinogenicity studies needed? 55515.4 Route and method of administration 55615.5 Selection of rodent species 55615.6 Setting of dose levels 55815.7 Group number and size 55915.8 Dietary considerations 56015.9 Time on study 56015.10 Organs to be evaluated 56115.11 Additional investigations 56215.12 Clinical trials and marketing times 56215.13 Clinical, autopsy findings and histopathological nomenclature . . 56315.14 Pathology technical issues 56415.15 Study data presentation and interpretation 56415.16 Discussion and conclusions 56515.17 References 566

16 Animal pharmacokinetics and toxicokinetics 569D. B. Campbell and R. Jochemsen

16.1 Introduction 56916.2 Regulatory requirements 57716.3 Discovery 57916.4 Species comparison of biodisposition 581

16.4.1 Regulatory requirements 58116.4.2 Analytical methods 58616.4.3 General considerations 58616.4.4 Absorption 58816.4.5 Distribution 589

16.4.5.1 Regulatory requirements 58916.4.5.2 Single dosing 58916.4.5.3 Repeated dosing 59116.4.5.4 Protein binding 59216.4.5.5 Foetal uptake 59316.4.5.6 Milk transfer 593

16.4.6 Metabolism 59616.4.6.1 Regulatory requirements 59616.4.6.2 Design 59616.4.6.3 Enzyme induction/inhibition 597

16.4.7 Elimination 59816.4.7.1 Mass balance 59816.4.7.2 Biliary elimination 599

16.4.8 Special animal populations 60316.5 Toxicokinetics 603

16.5.1 Regulatory requirements 603

Page 18: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xxi

16.5.2 Definition 60716.5.3 Studies 60716.5.4 Analytical methods 60716.5.5 Animals and dose groups 61016.5.6 Sampling 61016.5.7 Exposure measurement 61116.5.8 Determination of metabolites 61216.5.9 Complicating factors in exposure interpretation . . . . 61216.5.10 Acute dosing 61416.5.11 Repeat-dose toxicity 61416.5.12 Carcinogenicity 61516.5.13 Genotoxicity 61716.5.14 Reproductive and fertility studies 61716.5.15 Statistical evaluation 61916.5.16 Good Laboratory Practice 619

16.6 Stereoisomerism 61916.6.1 Regulatory requirements 61916.6.2 Single enantiomer 62216.6.3 Racemate 62216.6.4 Racemate-enantiomer switch 624

16.7 Expert Report 62416.8 The future 62516.9 Conclusions 62616.10 Acknowledgements 62716.11 References 627

PART III CLINICAL

17 Clinical pharmacology and pharmacodynamics 639A. J. Williams and D. T. Greenwood

17.1 Introduction 63917.2 The regulatory position 645

17.2.1 General requirements 64517.2.2 Specific therapeutic area/disease guidelines 648

17.3 References 649

18 Pharmacokinetics in man 650D. J. Nichols, P. E. Coates and D. A. Smith

18.1 Introduction 65018.2 Preclinical safety data 65118.3 Preclinical metabolism 65318.4 Bioanalytical methods 65418.5 Pharmacokinetic parameters 655

Page 19: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xxii Contents

18.6 Bioavailability and absorption parameters 65818.7 Pharmacokinetic-pharmacodynamic parameters 65818.8 Phase I studies 65918.9 Single-dose toleration and pharmacokinetics 66018.10 Single- and multiple-dose toleration and pharmacokinetics . . . . 66118.11 Dose proportionality: linearity and non-linearity with dose . . . . 66218.12 Bioavailability and bioequivalence 662

18.12.1 First bioavailability study 66318.12.2 Food effects on bioavailability 66318.12.3 Bioavailability of commercial dosage form 66418.12.4 Alternative oral formulations 66418.12.5 Alternative routes of administration 665

18.13 Metabolism 66518.14 Radiolabel biotransformation study 66618.15 Pharmacokinetics in special populations and situations 667

18.15.1 Drug interaction studies 66718.15.2 Pharmacokinetics in the elderly 66818.15.3 Pharmacokinetics in renal insufficiency 66818.15.4 Pharmacokinetics in hepatic insufficiency 669

18.16 Chiral drugs 66918.17 Population pharmacokinetics and pharmacodynamics 670

18.17.1 Phase I 67218.17.2 Phase Ha 67218.17.3 Phase lib 67218.17.4 Phase III 672

18.18 Integration: pharmacokinetics and pharmacodynamics 67218.19 Conclusions 67418.20 References 675

19 Ethnic, genetic and environmental factors 678L. P. Balant and O. Pelkonen

19.1 Introduction 67819.1.1 Pharmacogenetics 67819.1.2 Environmental factors 67919.1.3 Inter-ethnic differences in drug behaviour and action . . 68019.1.4 Scope of the chapter 680

19.2 Basic concepts and definitions 68119.2.1 Pharmacogenetics 68119.2.2 Environmental factors 68219.2.3 Inter-ethnic factors and pharmaco-anthropology . . . . 682

19.2.3.1 Inter-ethnic differences in drug behaviourand action 682

19.2.3.2 Pharmaco-anthropology 68319.2.4 Pharmacokinetics and pharmacodynamics 684

Page 20: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xxiii

19.3 Genetic factors 68419.3.1 Genetic polymorphism of N-acetylation 685

19.3.1.1 Historical and mechanistic considerations . . 68519.3.1.2 Clinical relevance 685

19.3.2 Genetic polymorphism of debrisoquine/sparteineoxidation 68619.3.2.1 Historical and mechanistic considerations . . 68619.3.2.2 Clinical relevance 686

19.3.3 Genetic polymorphism of mephenytoin oxidation . . . . 68719.3.3.1 Historical and mechanistic considerations . . 68719.3.3.2 Clinical relevance 687

19.3.4 Other polymorphisms 68719.3.5 Pharmacokinetic consequences 68819.3.6 Regulatory considerations for genetic polymorphisms . . 689

19.4 Environmental factors and enzymatic systems 69019.4.1 Relevant environmental factors 690

19.4.1.1 Cigarette smoking 69019.4.1.2 Alcohol drinking 69119.4.1.3 Concomitant drug therapy 69119.4.1.4 Occupational chemicals 69119.4.1.5 Other factors 692

19.4.2 Enzymatic systems in humans 69219.4.2.1 P450 enzymes 69219.4.2.2 Conjugative enzymes 695

19.5 Inter-ethnic and geographical factors 69519.5.1 Known genetic polymorphisms 696

19.5.1.1 N-Acetylation 69619.5.1.2 Debrisoquine/sparteine oxidation 69619.5.1.3 Mephenytoin oxidation 697

19.5.2 Nutrition 69719.5.2.1 Alcohol dehydrogenase 69819.5.2.2 Aldehyde dehydrogenase 698

19.5.3 Physical and chemical environment 69919.5.4 Pharmacodynamic aspects 699

19.6 Clinical study design and interpretation 70019.6.1 Pharmacokinetic/pharmacodynamic integration 700

19.6.1.1 Concepts and aims of PK/PD integration . . . 70019.6.1.2 Inter-ethnic differences in drug behaviour and

action and PK/PD integration 70119.6.2 Preclinical studies 701

19.6.2.1 Choice of animal species in toxicology . . . 70319.6.2.2 Animal models for environmental factors . . 703

19.6.3 Phase I studies 70519.6.3.1 Objectives and methodology 70519.6.3.2 Investigational pharmacokinetics 706

Page 21: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xxiv Contents

19.6.3.3 Bioavailability investigations 70619.6.3.4 Bioequivalence studies 70719.6.3.5 Investigational pharmacodynamics 70719.6.3.6 Studies in healthy volunteers for the detection

of inter-ethnic differences inpharmacokinetics 707

19.6.4 Phase II studies 70819.6.5 Phase IE studies 708

19.6.5.1 Methodological aspects in the context of theidentification of sources of variability in responseand/or behaviour 708

19.6.5.2 Studies in patients in the context of world-widemarketing 709

19.6.6 Phase IV studies 71019.6.6.1 Objectives and methodology 71019.6.6.2 Phase IV studies in the context of transcultural

drug licensing 71019.6.7 Genotyping, phenotyping and probe drugs 710

19.6.7.1 Genotyping for polymorphisms 71019.6.7.2 The concept of probe drugs 71119.6.7.3 Probe drugs for genetic polymorphisms . . . 71219.6.7.4 Probe drugs for environmental factors . . . . 71519.6.7.5 Cocktail approach 71819.6.7.6 Practical considerations for human studies . . 719

19.7 Regulatory considerations for transcultural registration 72019.8 Proposal for a strategy to discover and define sources of variability

from genetic and/or environmental origin 72119.9 Conclusions 72119.10 Acknowledgements 72219.11 References 722

20 Good Clinical Practice 737M. E. Allen

20.1 Introduction 73720.2 The basics of Good Clinical Practice 73820.3 Development of Good Clinical Practice in the United States . . . 739

20.3.1 Patients'rights 73920.3.2 Scientific misconduct and fraud 74120.3.3 Substantive and supportive data 742

20.4 Development of Good Clinical Practice outside the United States 74320.5 Differences in approach to Good Clinical Practice 74420.6 Legal status of Good Clinical Practice 745

20.6.1 The United States 74520.6.2 European Community 745

Page 22: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xxv

20.6.3 Japan 74620.7 Good Clinical Practice guidelines and regulations compared . . . 747

20.7.1 Ethics Committees and Institutional Review Boards . . 74720.7.1.1 The United States 74720.7.1.2 European Community 74820.7.1.3 Japan 749

20.7.2 Informed consent 75020.7.2.1 The United States 75020.7.2.2 European Community 75120.7.2.3 Japan 751

20.7.3 Obligations of investigators 75120.7.3.1 The United States 75220.7.3.2 European Community 75220.7.3.3 Japan 753

20.7.4 Obligations of sponsors and monitors 75520.7.4.1 The United States 75520.7.4.2 European Community 75620.7.4.3 Japan 758

20.7.5 Archiving of data 75820.7.5.1 The United States 75820.7.5.2 European Community 75920.7.5.3 Japan 759

20.7.6 Audit of sponsors and authorities 75920.7.6.1 The United States 75920.7.6.2 European Community 76020.7.6.3 Japan 761

20.8 Harmonisation of Good Clinical Practices 76120.9 Good Clinical Practice legislation and guidelines 762

20.9.1 The United States 76220.9.1.1 Regulations 76220.9.1.2 Guideline 76220.9.1.3 Compliance program Guidance Manuals . . 76220.9.1.4 Guidance 762

20.9.2 European Community 76320.9.2.1 Guideline 76320.9.2.2 Other legal and technical requirements . . . . 763

20.9.3 Japan 76 ̂20.9.3.1 Legal requirements "h320.9.3.2 Guideline 763

20.10 Acknowledgement 76420.11 References 7 M

Page 23: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xxvi Contents

21 Clinical trials - general aspects of design and interpretation 765J. Dahlstrom, C. R. Griffett and C. M. Perkins

21.1 Introduction 765

21.2 Aspects of design of clinical trials 766

21.2.1 Experimental design on the basis of disease type . . . . 76621.2.2 Types of clinical trial 768

21.2.2.1 Definition 76821.2.2.2 Phase I 76921.2.2.3 Phase II 76921.2.2.4 Phase HI 76921.2.2.5 Phase IV 769

21.2.3 Controlled/uncontrolled trial design 77021.2.3.1 Controlled trials 77021.2.3.2 Uncontrolled trials 770

21.2.4 Parallel group or cross-over design? 770

21.3 Specific features of clinical trial design and interpretation . . . . 77121.3.1 The protocol 77121.3.2 Objectives of the trial 77221.3.3 Study size 772

21.3.3.1 Statistical considerations 77321.3.4 Patient selection 77421.3.5 Randomisation 77521.3.6 Blinding 77621.3.7 Drug dosage 77621.3.8 Duration of Study 777

21.4 Clinical trial interpretation 77721.4.1 Outcome measures 77721.4.2 Interim monitoring 77821.4.3 Concomitant medication 77921.4.4 Patient compliance 77921.4.5 Withdrawals 77921.4.6 Multinational clinical trials 780

21.5 The impact of Good Clinical Practice on clinical trial design andinterpretation 780

21.5.1 Designing a clinical trial in accord with GCP 781

21.5.2 Ethical Committees/Institutional Review Boards . . . . 78121.5.3 Informed consent 781

21.5.4 Standard Operating Procedures/Quality assurance . . . 78221.6 Future directions 78321.7 References 78421.8 Guidelines I 78521.9 Guidelines II 786

Page 24: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xxvii

22 Statistical analysis of clinical data 787R. T. O'Neill

22.1 Introduction 78722.2 Statistical principles 788

22.2.1 The current status of published documents describingstatistical principles in medical and pharmaceuticalresearch 78822.2.1.1 United States 78922.2.1.2 Nordic countries 79022.2.1.3 European Community 79122.2.1.4 Japan 791

22.2.2 Other relevant published literature 79222.3 Some analysis issues 793

22.3.1 Randomisation: assessing whether the goal was achieved 79422.3.2 Statistical adjustments for co-variates 79522.3.3 Subgroup analysis, patient subsets and variation in

therapeutic efficacy or safety 79622.3.3.1 When are the subgroups determined? . . . . 799

22.3.4 Intent to treat analysis of clinical trials with respect toprotocol violations and missing data 79922.3.4.1 The purpose of a trial: Phase II and Phase III

trials 80022.3.4.2 Some FDA guidance as stated in the guidelines . 80022.3.4.3 Operational questions 801

22.3.5 Compliance and adherence to drug assignment inclinical trials 801

22.3.6 Statistical analysis of longitudinally collected data,patient dropouts, and differential patient follow-upand exposure 802

22.3.7 Sequential and group sequential clinical trials: Interimanalysis and unplanned analyses of accruing data . . . . 804

22.3.8 Cross-over designs and their analysis 80722.3.9 Multicentre studies 809

22.3.9.1 Interpreting the variability in treatment effectsamong centres 810

22.3.9.2 Methods of analysis HI 122.3.9.3 Multinational multicentre studies 811

22.3.10 Active control trials: claiming treatment effects areequivalent to those of another effective agent 812

22.3.11 The analysis of dose-response: dose^ranging studies . . 81422.3.12 Factorial designs 81522.3.13 Fixed dose combination drug designs 816

22.3.13.1 Should a placebo group be included in the fixedcombination trial? 817

Page 25: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

xxviii Contents

22.3.14 Meta-analysis and systematic overviews of clinicalstudies in licence applications 817

22.3.15 The statistical analysis of safety outcomes in clinicalstudies 819

22.4 Summary and concluding remarks 82022.5 References 821

PART IV BIOLOGICAL PRODUCTS AND BIOTECHNOLOGY

23 Clinical aspects of recombinant DNA products 833F. Rotblat

23.1 Introduction 83323.2 Pharmacodynamics 83323.3 Pharmacokinetics 83423.4 Efficacy 83523.5 Replacement proteins 83523.6 Pharmacological overdoses 83623.7 Cytokines and interferons 83623.8 Products for the future 83723.9 Tailoring the indication 83723.10 Safety 83723.11 Safety analysis 83723.12 Problems specific to recombinant products 838

23.12.1 Secondary pharmacology 83823.12.2 Antibodies 83823.12.3 Transmission of viral infection 83823.12.4 Residual DNA 83823.12.5 Contaminants from the manufacturing processes . . . . 83923.12.6 Changes in structure 839

23.13 Conclusions 83923.14 References 839

24 Preclinical pharmacological and toxicological requirements forbiological and biotechnological products 840R. M. Lee

24.1 Introduction 84024.2 Toxicology 841

24.2.1 Single-dose toxicity 84124.2.2 Repeated-dose toxicity 84224.2.3 Reproduction toxicity 84324.2.4 Mutagenic potential 84524.2.5 Carcinogenic potential 84624.2.6 Local tolerance 846

24.3 Pharmacology 846

Page 26: INTERNATIONAL PHARMACEUTICAL PRODUCT REGISTRATION · 7.2.4 Kinetics of chemical degradation reactions in pharmaceutical products 212 7.2.5 Chemical stability of solids 213 7.2.6 Toxicity

Contents xxix

24.3.1 Pharmacology (I): related to the desired action 84624.3.2 Pharmacology (II): not related to the desired action . . . 846

24.4 Pharmacokinetics and biotransformation 84724.5 References 847

24.5.1 General 84824.5.2 Specific guidance 848

Annex 1 - List of the principal abbreviations and acronyms used in thetext 849Annex 2 — Information sources on legal and technical requirements forregistration of medicinal products in the EC, Japan and the United States 856

Index 872