regulated bioanalytical...

30
REGULATED BIOANALYTICAL LABORATORIES Technical and Regulatory Aspects from Global Perspectives MICHAEL ZHOU Ph.D. Synta Pharmaceuticals Corporation

Upload: doanquynh

Post on 21-Jun-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

REGULATEDBIOANALYTICALLABORATORIES

Technical and Regulatory Aspectsfrom Global Perspectives

MICHAEL ZHOU Ph.D.

Synta Pharmaceuticals Corporation

Page 2: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1
Page 3: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

REGULATED BIOANALYTICAL LABORATORIES

Page 4: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1
Page 5: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

REGULATEDBIOANALYTICALLABORATORIES

Technical and Regulatory Aspectsfrom Global Perspectives

MICHAEL ZHOU Ph.D.

Synta Pharmaceuticals Corporation

Page 6: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

Copyright � 2011 by John Wiley & Sons, Inc. All rights reserved

Published by John Wiley & Sons, Inc., Hoboken, New Jersey

Published simultaneously in Canada

No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by

any means, electronic, mechanical, photocopying, recording, scanning, or otherwise, except as permitted

under Section 107 or 108 of the 1976 United States Copyright Act, without either the prior written

permission of the Publisher, or authorization through payment of the appropriate per-copy fee to the

Copyright Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, (978) 750-8400, fax (978)

750-4470, or on the web at www.copyright.com. Requests to the Publisher for permission should be

addressed to the Permissions Department, JohnWiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030,

(201) 748-6011, fax (201) 748-6008, or online at http://www.wiley.com/go/permission.

Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts in

preparing this book, they make no representations or warranties with respect to the accuracy or

completeness of the contents of this book and specifically disclaim any implied warranties of

merchantability or fitness for a particular purpose. No warranty may be created or extended by sales

representatives orwritten salesmaterials. The advice and strategies contained hereinmay not be suitable for

your situation. You should consult with a professional where appropriate. Neither the publisher nor author

shall be liable for any loss of profit or any other commercial damages, including but not limited to special,

incidental, consequential, or other damages.

For general information on our other products and services or for technical support, please contact our

Customer Care Department within the United States at (800) 762-2974, outside the United States at (317)

572-3993 or fax (317) 572-4002.

Wiley also publishes its books in a variety of electronic formats. Some content that appears in print may not

be available in electronic formats. For more information about Wiley products, visit our web site at www.

wiley.com.

Library of Congress Cataloging-in-Publication Data:

Zhou, Michael.

Regulated bioanalytical laboratories : technical and regulatory aspects from global

perspectives / Michael Zhou.

p. cm.

Includes index.

ISBN 978-0-470-47659-8 (cloth)

1. Medical laboratories–Qualtiy control. 2. Biological laboratories–Qualtiy control.

3. Pharmaceutical technology–Qualtiy control. I. Title.

R860.Z56 2011

610.28’4–dc22

2010022398

Printed in United States of America

10 9 8 7 6 5 4 3 2 1

Page 7: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

CONTENTS

Preface xiii

Acknowledgment xvii

Contributors and Advisors xix

1 Introduction, Objectives, and Key Requirements

for GLP Regulations 1

1.1 Introduction 1

1.1.1 Good Laboratory Practices 1

1.1.2 Bioanalytical Laboratories—Bioanalysis 4

1.1.3 Good Laboratory Practices Versus Bioanalytical Labs/

Bioanalysis 7

1.2 Objectives and Key Requirements for GLP Regulations 8

1.3 Fundamental Understanding of GLP Regulations and Principles 10

1.3.1 Elements of Good Laboratory Practices 11

1.4 Key Elements of Bioanalytical Methods Validation 16

1.4.1 Reference Standards 19

1.4.2 Method Development—Chemical/Chromatographic

Assay 20

1.4.3 Calibration/Standard Curve 21

1.4.4 Stability 21

1.4.5 Reproducibility 23

1.4.6 Robustness or Ruggedness 23

v

Page 8: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

1.5 Basic Principles of Bioanalytical Method Validation and

Establishment 23

1.5.1 Specific Recommendations for Method Validation 24

1.5.2 Acceptance Criteria for Analytical Run 29

References 33

2 Historic Perspectives of GLP Regulations, Applicability,

and Relation to Other Regulations 35

2.1 Historic Perspectives of GLP Regulations 35

2.1.1 Economic Assessment 39

2.1.2 Environmental Impact 40

2.2 Applicability and Relations to Other Regulations/Principles 42

2.2.1 GLP, GCP, GMP, and Part 11 42

2.2.2 General Terminologies and Definitions of GxPs (GLP,

GCP, and cGMP) 47

2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD

GLP Principles 47

2.3.1 US and OECD GLP Similarity and Differences 53

2.4 Applications of GLP to Multiple Site Studies 55

2.4.1 Roles and Responsibilities 57

2.4.2 Performance of the Studies 61

2.4.3 Applications of GLP to In Vitro Studies for Regulatory

Submissions 64

2.5 21 CFR Part 11 in Relation to GLP Programs 66

2.5.1 A New Risk-Based Approach 67

2.5.2 Understanding Predicate Rule Requirements 67

2.5.3 21 CFR Part 11 Best Practices 68

2.5.4 Use of Electronic Signatures 71

2.6 GLP, cGMP, and ISOApplicabilities, Similarity, and Differences 74

2.6.1 GLPs, cGMPs, ISO 17025:2005: How Do They Differ? 74

2.6.2 GLPs Versus GMPs 74

2.6.3 GLPs Versus ISO/IEC 17025:2005 75

2.6.4 ISO Versus GLPs 76

2.7 Good Clinical Practices and Good Clinical Laboratory Practices 78

2.8 Gap andCurrent Initiatives onRegulatingLaboratoryAnalysis in

Support of Clinical Trials 80

References 84

3 GLP Quality System and Implementation 87

3.1 GLP Quality System 87

3.1.1 Regulatory Inspection for GLP Quality System 95

3.1.2 Good Laboratory Practice Inspections 99

3.1.3 GLP Quality System Objectives 103

3.2 Global GLP Regulations and Principles 106

3.2.1 General 106

vi CONTENTS

Page 9: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

3.2.2 Responsibilities and Compliance 107

3.2.3 Statement of Compliance in the Final Report 107

3.2.4 Protocol Approval 108

3.2.5 Assignment of Study Director 108

3.2.6 Laboratory Qualification/Certification 108

3.2.7 Authority Inspections 108

3.2.8 Archiving Requirements 108

3.3 Implementation of GLP Regulations and OECD Principles 109

3.3.1 Planning (Master Schedule) 114

3.3.2 Personnel Organization 115

3.3.3 Curriculum Vitae 115

3.3.4 Rules of the Conducts of Studies 116

3.3.5 Content of Study Protocol 116

3.3.6 Approval of Study Protocol 118

3.3.7 Distribution of Study Protocol 118

3.3.8 Protocol Amendment 118

3.3.9 Standard Operating Procedures 119

3.3.10 SOP System Overview 119

3.3.11 Characterization 121

3.3.12 Test Item/Article Control before Formulation 121

3.3.13 Preparation of the Dose Formulation 123

3.3.14 Sampling and Quality Control of Dose Formulation 125

3.4 Initiatives and Implementation of Bioanalytical Method

Validation (Guidance for Industry BMV—May 2001) 126

3.4.1 Summary 127

References 128

4 Fundamental Elements and Structures for Regulated

Bioanalytical Laboratories 131

4.1 Introduction 131

4.2 Fundamental Elements for Bioanalytical Laboratories 133

4.2.1 Document Retention and Archiving 136

4.3 Basic Requirements for GLP Infrastructure and Operations 139

4.4 GxP Quality Systems 143

4.4.1 Laboratory Instrument Qualification and Validation 149

4.4.2 Procedural Elements and Function that Maintain

Bioanalytical Data Integrity for GLP Studies 150

References 166

5 Technical and Regulatory Aspects of Bioanalytical Laboratories 167

5.1 Fundamental Roles and Responsibilities of

Bioanalytical Laboratories 167

5.1.1 Technical Functions of Bioanalytical Laboratories 168

5.1.2 Basic Processes in Bioanalytical Method Development,

Validation, and Sample Analysis 173

CONTENTS vii

Page 10: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

5.2 Qualification of Personnel, Instrumentation, and

Analytical Procedures 178

5.2.1 FromRegulatory Perspectives: Personnel, Training, and

Qualification 183

5.2.2 Facility Design and Qualifications 186

5.2.3 Equipment Design and Qualification 186

5.2.4 Analytical/Bioanalytical Method Qualification and

Validation along with Related SOPs 197

5.3 Regulatory Compliance with GLP Within

Bioanalytical Laboratories 204

5.4 Joint-Effort from Industries and Regulatory Agencies 206

5.4.1 Ligand-Binding Assays In-Study Acceptance Criteria 213

5.4.2 Determination ofMetabolites during Drug Development 216

5.4.3 Incurred Sample Analysis 216

5.4.4 Documentation Issues 217

5.4.5 Analytical/Validation Reports 218

5.4.6 Source Data Documentation 218

5.4.7 Final Report Documentation 219

5.4.8 Stability Recommendation 219

5.4.9 Matrix Effects for Mass Spectrometric-Based Assays 221

5.4.10 System Suitability 222

5.4.11 Reference Standards 222

5.4.12 Validation Topics with No Consensus 222

5.4.13 Specific Criteria for Cross-Validation 223

5.4.14 Separate Stability Experiments Required at � 70�C if

Stability Shown at � 20�C 223

5.4.15 Stability Criteria for Stock Solution Stability 224

5.4.16 Acceptance Criteria for Internal Standards 224

5.4.17 Summary 224

References 226

6 Competitiveness of Bioanalytical Laboratories—Technical

and Regulatory Perspectives 229

6.1 Technical Aspect of Competitive Bioanalytical Laboratories 229

6.2 Bioanalytical Processes and Techniques 232

6.2.1 Sample Generation, Shipment, and Storage 232

6.2.2 Sample Preparation 233

6.3 Enhancing Throughput and Efficiency in Bioanalysis 243

6.3.1 Chromatographic Separation 244

6.3.2 Selective and Sensitive Detection 251

6.4 Technical Challenges and Issues on Regulated Bioanalysis 254

6.4.1 Matrix Effect 254

6.4.2 Method Validation and Critical Issues during Sample

Analysis 256

6.4.3 Method Transfer 258

viii CONTENTS

Page 11: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

6.5 Regulatory Aspects of Competitive Bioanalytical Laboratories 264

6.5.1 General Consideration 264

6.5.2 Historical Perspective 265

6.5.3 Personnel—Training and Qualification 267

6.5.4 Facility—Design and Qualifications 269

6.5.5 Equipment Design and Qualification 270

6.5.6 Standard Operating Procedures 272

6.5.7 Laboratory/Facility Qualification Perspectives 272

6.6 Advanced/Competitive Bioanalytical Laboratories 277

6.6.1 Strategy Versus Tactics 278

6.6.2 Bioanalytical Laboratory Assessment 279

6.6.3 Capacity 279

6.6.4 Experience 280

6.6.5 Quality 281

6.6.6 Performance and Productivity Measures 281

6.6.7 Information Technology and Data Management 282

6.6.8 Communication 282

6.6.9 Financial Stability 283

6.6.10 Ease of Use 283

6.6.11 Contracting Bioanalytical Services 284

6.6.12 The Contracting Process 284

6.7 Applications andAdvances in Biomarker and/or Ligand-Binding

Assays within Bioanalytical Laboratories 286

References 290

7 Sponsor and FDA/Regulatory Agency GLP Inspections and Study

Audits 297

7.1 GLP versus Biomedical Research Monitoring and Mutual

Acceptance of Data for Global Regulations and Inspections 298

7.2 Purposes and Benefits of Regulatory Inspections/Audits 303

7.2.1 Criteria for Selecting Ongoing and Completed Studies 304

7.2.2 Areas of Expertise of the Facility 305

7.2.3 Establishment Inspections 305

7.2.4 Organization and Personnel (21 CFR 58.29, 58.31,

58.33) 305

7.2.5 Quality Assurance Unit (QAU; 21 CFR 58.35) 307

7.2.6 Facilities (21 CFR 58.41–58.51) 308

7.2.7 Equipment (21 CFR 58.61–58.63) 309

7.2.8 Testing Facility Operations (21 CFR 58.81) 310

7.2.9 Reagents and Solutions (21 CFR 58.83) 311

7.2.10 Animal Care (21 CFR 58.90) 311

7.2.11 Test and Control Articles (21 CFR 58.105–58.113) 312

7.2.12 Test and Control Article Handling (21 CFR 58.107) 313

7.2.13 Protocol and Conduct of Nonclinical Laboratory Study

(21 CFR 58.120–58.130) 314

CONTENTS ix

Page 12: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

7.2.14 Study Protocol (21 CFR 58.120) 314

7.2.15 Test System Monitoring 314

7.2.16 Records and Reports (21 CFR 58.185–58.195) 314

7.2.17 Data Audit 316

7.2.18 General 316

7.2.19 Final Report Versus Raw Data 317

7.2.20 Specimens Versus Final Report 318

7.2.21 Refusal to Permit Inspection 318

7.2.22 Sealing of Research Records 318

7.2.23 Samples 319

7.3 Typical Inspections/Audits and Their Observations 320

7.4 Regulatory Challenges for Bioanalytical Laboratories 321

7.4.1 Introduction 321

7.4.2 Analysis of Current FDA Inspection Trends 324

7.4.3 Discussion and Analysis of Specific Potential FDA 483

Observation Issues 325

7.4.4 Method Validation Issues 325

7.4.5 Batch Runs Acceptance Criteria Issues 329

7.4.6 Events/Deviations Investigation/Resolution Issues 331

7.4.7 Test Specimen Accountability Issue 333

7.4.8 Recommendations to Support an Effective FDA

Inspection Readiness Preparation 334

7.5 Handling and Facilitating GLP or GxP Audits/

Inspections 334

7.5.1 General Preparation for an Inspection 336

7.5.2 Why Are Audits/Inspections Needed and Conducted? 342

7.5.3 Written Policy in Place 342

7.5.4 Positions on Controversial Issues 343

7.5.5 The Inspection Coordinator 344

7.5.6 Follow-Up Procedures 348

7.5.7 Summary 349

References 351

8 Current Strategies and Future Trends 353

8.1 Strategies from General Laboratory and Regulatory

Perspectives 354

8.2 Strategies from Technical and Operational Perspectives 356

8.3 Biological Sample Collection, Storage, and Preparation 360

8.3.1 Sample Collection and Storage 360

8.3.2 Sample Preparation Techniques 361

8.3.3 Off-Line Sample Extraction 364

8.3.4 On-Line Sample Extraction 364

8.4 Strategies for Enhancing Mass Spectrometric Detection 366

8.4.1 Enhanced Mass Resolution 368

8.4.2 Atmospheric Pressure Photoionization 369

x CONTENTS

Page 13: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

8.4.3 High-Field Asymmetric Waveform Ion Mobility

Spectrometry 370

8.4.4 Electron Capture Atmospheric Pressure Chemical

Ionization 370

8.4.5 Mobile Phase Optimization for Improved Detection and

Quantitation 371

8.4.6 Anionic and Cationic Adducts as Analytical Precursor

Ions 372

8.4.7 Derivatization 372

8.5 Strategies for Enhancing Chromatography 374

8.5.1 Ultra-Performance Chromatography 375

8.5.2 Hydrophilic Interaction Chromatography for Polar

Analytes 376

8.5.3 Specialized Reversed-Phase Columns for Polar

Analytes 377

8.5.4 Ion-Pair Reversed-Phase Chromatography for Polar

Analytes 378

8.6 Potential Pitfalls in LC–MS/MS Bioanalysis 378

8.6.1 Interference from Metabolites or Prodrugs due to

In-Source Conversion to Drug 378

8.6.2 Interference from Metabolites or Prodrugs due to

Simultaneous M þ Hþ and M þ NH4þ Formation or

Arising from Isotopic Distribution 379

8.6.3 Pitfall in Analysis of Two Interconverting Analytes due

to Inappropriate Method Design 383

8.6.4 Matrix Effect 383

8.7 Trends in High-Throughput Quantitation 386

8.7.1 System Throughput 386

8.7.2 High-Speed HPLC 386

8.8 Trends in Hybrid Coupling Detection Techniques 388

8.9 Trends in Internal R&D and External Outsourcing 388

8.10 Trends in Ligand-Binding Assays and LC–MS/MS

for Biomarker Assay Applications 397

8.11 Trends in Study Design and Evaluation

Relating to Bioanalysis 399

8.12 Trends in Applying GLP to In Vitro Studies in Support of

Regulatory Submissions 403

8.13 Trends in Global R&D Operations 404

8.14 Trends in Regulatory Implementations 407

8.14.1 Calibration Range and Quality Control Samples 407

8.14.2 Incurred Sample Reproducibility (Duplicate Sample

Analysis) 408

8.14.3 LIMS and Electronic Data Handling, Security,

Archiving, and Submission 409

8.15 Trends in Global Regulations and Quality Standards 412

CONTENTS xi

Page 14: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

8.16 Trends in Compliance with 21 CFR Part 11 414

8.16.1 21CFR Part 11 Software Requirements 415

8.16.2 Building a Roadmap for Compliance with

21 CFR Part 11 415

8.16.3 Low Hanging Fruits in the Roadmap for Compliance

with 21 CFR Part 11 416

8.17 Summary 419

References 421

9 General Terminologies of GxP and Bioanalytical Laboratories 431

9.1 General Terminologies for GxP and Bioanalytical Laboratories 431

9.2 GLP Basic Concepts and Implementation 469

9.2.1 The Study Protocol 470

9.2.2 Raw Data 471

9.2.3 The GLP Archive and the Archivist 472

9.2.4 Expansion of GLP Scope 473

9.2.5 OECD GLP 473

9.3 GLP Guidance Documents 474

9.3.1 FDA Guidance for Industry on Bioanalytical Method

Validation 474

9.3.2 OECD GLP Guidance Documents 474

9.3.3 Swiss GLP Guidance Documents 475

References and Sources for Above Terminologies 475

Appendix A Generic Checklist for GLP/GXP Inspections/Audits 479

Appendix B General Template for SOP 489

Appendix C Typical SOPs for GLP/Regulated Bioanalytical

Laboratory 493

Quality Assurance—GLP 493

Bioanalytical—GLP Laboratories 494

Appendix D Basic Equipment/Apparatus for Bioanalytical

Laboratory 497

Appendix E Website Linkages for Regulated Bioanalysis 499

Index 503

xii CONTENTS

Page 15: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

PREFACE

TheGood Laboratory Practice (GLP) regulations were established in the 1970s by the

United States Food and Drug Administration (FDA), and published in the Code of

Federal Regulations (21 CFR Part 58). The Organization for Economic Cooperation

and Development (OECD) established the Principles of GLP in 1981. United States

Environmental Protection Agency (EPA) adopted its own set of GLP regulations

shortly thereafter, governing the research surrounding pesticides and toxic chemicals.

Bioanalytical laboratories have increasingly become critically important in data

generation for discovery, preclinical, and clinical development in life science

industries. Bioanalysis, employed for the quantitative determination of drugs and

their metabolites in biological fluids, plays significant roles in the evaluation and

interpretation of bioequivalence (BE), bioavailability (BA), pharmacodynamic (PD),

pharmacokinetic (PK), and toxicokinetic (TK) studies. The quality of these studies,

which are often used to support regulatory filings, is directly related to the quality of

the underlying bioanalytical data. It is therefore important that guiding principles for

the validation of these analytical methods be established and disseminated to the

pharmaceutical and life science communities.

The focus and working groups from American Association of Pharmaceutical

Scientists (AAPS), European Bioanalysis Forum (EBF), Food and Drug Adminis-

tration, European Medicine Agency (EMEA), and other related organizations have

held a series of workshops and seminars focusing on key issues relevant to bioana-

lytical methodology and provided a platform for scientific discussions and delibera-

tions. As bioanalytical tools and techniques have continued to evolve and significant

scientific and regulatory experiences have been gained, the bioanalytical community

has continued its critical review of the scope, applicability, and success of the

presently employed bioanalytical guiding principles. Life science products including

xiii

Page 16: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

foods, nutritional supplements, medicine/drug discovery, research, and development

expand to wide scope of life sciences meaning varieties of industries such as

environmental toxicology, food nutritional analysis, biotechnology, biopharmaceu-

ticals, pharmaceuticals, hospitals, diagnostic/medical device industries, and so on.

Bioanalytical sciences basically support above-mentioned areas under FDAGLP and

other related regulations, principles, and guidelines. Below are some example

elements/infrastructure that are required for regulated Bioanalytical Laboratories:

(1) Responsible management team with quality system

(2) Qualified personnel selection, staffing, and training

(3) Standard operating procedures (SOPs)

(4) Installation, operational, and performance qualification (IQ, OQ, and PQ,

respectively) of facilities, instrumentation, and software

(5) Quality control (QC) procedures and staffing

(6) Quality assurance unit (QAU)

(7) Data generation and security assessment

(8) Documentation and archival process

(9) Laboratory information management system (LIMS)

(10) Final gap analysis

This book provides useful information for bioanalytical/analytical scientists,

analysts, quality assurance managers, and all personnel in bioanalytical laboratories

through all aspects of bioanalytical technical and regulatory perspectives within

bioanalytical operations and processes. Readers will learn how to develop and

implement strategies for routine, nonroutine, and standard bioanalytical methods

and on the entire equipment hardware and software qualification process. The book

also gives guidelines on qualification of certified standards and in-house reference

material aswell as on people qualification. Finally, it guides readers through stressless

internal and third party laboratory audits and inspections. Highly comprehensive

content with specific chapter by chapter is elaborated making it easy not only to learn

the subject but also to quickly implement the recommendations.

It takes account to most national and international regulations and quality and

accreditation standards such as GLP, cGMP, GCP, and GCLP from US FDA, ICH,

WHO, and EU, accreditation standards such as ISO17025 and to corresponding

interpretation and inspection guides. The text beginswith an introductory overview of

the roles of bioanalytical laboratories in pharmaceutical and biotechnology drug

development. Regulatory wise, it describes some fundamental understanding of

regulatory aspectswithin bioanalytical laboratories—current and future requirements

as far as GLP and/or GxP quality systems, facility, and personnel infrastructure and

qualification along with continuing improvement on a daily basis. From technical

standpoints, the book also elaborates the strategies for sample preparation, along with

essential concepts in extraction chemistry. Particular strategies for efficient use of

automation within bioanalytical laboratories are also presented. With regards to

xiv PREFACE

Page 17: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

instrumental analysis, fundamental approach is presentedwithin the areas of LC–MS/

MS and other hyphenated analytical techniques. Ligand-binding assays are also

discussed to recognize its increasingly crucial applications within bioanalytical

laboratories. Important objectives that can be accomplished when the strategies

presented in this book are followed include: improved efficiency inmoving discovery

compounds to nonclinical and clinical status with robust analytical methods; auto-

mation for sample preparation; modern analytical equipment, and improved knowl-

edge and expertise of laboratory staff. It has been widely accepted that good sciences

are not enough to meet regulatory requirements. In author�s opinion, good laboratorypractices may not improve any “poor” sciences, but indeed make “good sciences”

better as to ensure the quality, integrity, and reconstructability of data. GLP or GxP is

all about documentation. In anotherword, nothing has been properly donewithout any

documentation. GLP principles may also enhance the opportunity of Limiting waste

of resources, Ensuring high quality of data, Acquiring comparability of results, and

Deriving to mutual recognition of scientific findings worldwide and ultimately

Securing the health and well-being of our societies, as being LEADS concept per

author�s perspectives.

PREFACE xv

Page 18: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1
Page 19: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

ACKNOWLEDGMENT

The need of this book has been apparent as noted that Good Laboratory Practice must

be followed while generating data for regulatory consideration. I am grateful to peer-

reviewers for their positive feedback and encouragement since this project started.

The staff at John Wiley and Sons has provided me with tremendous support. In

particular, I would like to thankmy editor, Jonathan Rose, who has been an invaluable

resource during the project as well as other advice. I am truly indebted to all of the

contributors for their willingness of sharing their experiences, knowledge, and

perspectives on bioanalytical technical and regulatory aspects. The contributions as

well as the many discussions and interactions are worth noting! My special gratitudes

extend to Dr. Vinod P. Shah for his expert advice and inputs. The growth of

bioanalytical laboratory operations and contributions in life science industries has

been truly remarkable. I am thankful to have had the opportunity to interact with so

many people (contributors, reviewers, and advisors) who shared a common passion

for the analytical/bioanalytical sciences and regulatory compliance for the betterment

of the world and health.

Finally, I thank my wife, family, and friends for their courage and continued

support for everything I do.

xvii

Page 20: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1
Page 21: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

CONTRIBUTORS AND ADVISORS

Author sincerely appreciates all of the advice, review, suggestions, and edits from

following field experts on respective chapters and sections.

Frank Chow Lachman Consultants

1600 Stewart Avenue, Suite 604

Westbury, NY 11590, USA

Howard M. Hill Huntingdon Life Sciences

Huntingdon, Cambridge, PE28 4HS, UK

Mohammed Jemal Bristol-Myers Squibb

Route 206 and Province Line Road

Princeton, NJ 08543, USA

Marian Kelley MKelley Consulting LLC

1533 Glenmont Lane

West Chester, PA 19380, USA

Jean Lee Amgen Inc

100 Amgen Center Drive

Thousand Oaks, CA 91320, USA

Raymond Naxing Xu Abbott Laboratories

100 Abbott Park Road

Abbott Park, IL 60064, USA

xix

Page 22: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1
Page 23: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

1INTRODUCTION, OBJECTIVES,AND KEY REQUIREMENTSFOR GLP REGULATIONS

1.1 INTRODUCTION

1.1.1 Good Laboratory Practices

Good laboratory practices (GLPs 21 CFR PART 58) is a standard by which laboratory

studies are designed, implemented, and reported to assure the public that the results

are accurate/reliable and the experiment can be reproduced accordingly [1], at any

time in the future. In less technical terms, GLP is the cornerstone of all laboratory-

based activities in any organization that prides itself on the quality of the work it

performs. And, despite its immediate association with the pharmaceutical sector,

GLPs can (and should) be applied to virtually all industries in which laboratory work

is conducted, including companies involved in drug development, manufacturing,

foods, pesticides (agrochemicals), drink production, and engineering testing. In

addition, commercial testing laboratories (for toxicology, metabolism, materials,

and safety, for example), research establishments, and universities—in fact, all

laboratories engaged in product or safety testing or research and development—

should adopt and apply the doctrines of GLP.

GLP is not a luxury. It is a necessity for any professional laboratorywishing to gain

and retain the respect of its employees, clients, regulators, and perhaps most

importantly, its competitors. If a company is seen to be applying and adhering to

the highest standards of laboratory practice, it will gain significant competitive

advantage and will compete successfully for business and recognition within its

Regulated Bioanalytical Laboratories: Technical and Regulatory Aspects from Global Perspectives,

By Michael Zhou

Copyright � 2011 John Wiley & Sons, Inc.

1

Page 24: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

operational environment. Conversely, without rigidly enforced GLPs, good clinical

practice (GCP) [2], good manufacturing practices (GMPs) [3], or GxPs—a scientific

organizationwill not achieve the commercial success and respect that its products and

personnel deserve.

Published GLP regulations and guidelines have a significant impact on the daily

operations of analytical and/or bioanalytical laboratories. GLP is a regulation that

enhances good analytical practice. Good analytical/bioanalytical practice is impor-

tant, but it is not enough. For example, the laboratory must have a specific

organizational structure and procedures to perform and document laboratory work.

The objective is not only quality of data but also traceability and integrity of data.

However, the biggest difference between GLP and non-GLP work is the type and

amount of documentation. GLP functions as a regulation, which deals with the

specific organizational structure and documents related to laboratory work in order to

maintain integrity and confidentiality of the data. The entire cost of GLP-based work

is about 40% or more additional (from case to case) when compared to non-GLP

operations. For aGLP inspector, it should be possible to look at the documentation and

to easily find out the following:

. Who has done a study

. How the experiment was carried out

. Which procedures have been used, and

. Whether there has been any problem and if so

. How it has been addressed and solved where applicable

And this should not only be possible during and right after the study has been

finished but also 5–10 or more years later.

From worldwide perspectives, good practice rules govern drug/product develop-

ment activities in many parts of theworld.World Health Organization (WHO), which

has published documents on current good manufacturing practices (cGMPs) and

GCPs, has not previously recommended or endorsed any quality standard governing

the nonclinical phases of drug/product development. GLPs are recognized rules

governing the conduct of nonclinical safety studies, ensuring the quality, integrity, and

reliability of their data. To introduce the concepts of GLP to scientists in developing

countries, workshops onGLP have been organized in these regions. As an outcome of

the workshops (industries and regulatory bodies), it became apparent that some

formal guidance would be needed for the successful implementation of the GLP

regulations.

The first scientific working group on GLP issues was convened on November 25,

1999, in Geneva, to discuss quality issues in general and the necessity for a WHO

guidance document on GLP in particular. The working group concluded that it was

important to avoid the coexistence of two GLP standards, the Principles of good

laboratory practice of the Organization for Economic Cooperation and Development

(OECD) [4] being the internationally recognized and accepted standard, and

recommended that theOECDPrinciples be adopted byWHOforResearch&Training

2 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

Page 25: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

in Tropical Disease (TDR) as the basis of this guidance document. The experts also

recognized the need to address quality issues in areas other than the strictly regulated

safety studies for regulatory submission, and recommended that some explanation be

included in this guidance document. The working group further recommended that

WHO/TDR should request OECD’s permission to publish the existing OECD GLP

textwith aWHOendorsement, and to supplement it with an explanatory introduction.

Classical drug development (drug life cycle) is characterized by four well-defined

stages as follows:

Stage 1: The first stage, the discovery of potential new drug products, is neither

covered by a regulatory standard, nor are studies demonstrating proof of

concept. This area may well require some international standards or guidance

documents in the future.

Stage 2: The position of GLP studies within the drug development process is

specific to the second stage. These studies are termed “nonclinical” as they are

not performed in human. Their primary purpose is safety testing. Toxicology

and safety pharmacology studies, with a potential extension to pharmacoki-

netics and bioavailability, are those studies where the compliance with GLP is

required, which is the rather restricted scope of GLP.

Stage 3: The third stage, following on from safety studies, encompasses the clinical

studies in human. Here, GCP is the basis for quality standards, ethical conduct,

and regulatory compliance. GCP must be instituted in all clinical trials from

Phase I (to demonstrate tolerance of the test drug and to define human

pharmacokinetics), through Phase II (where the dose–effect relationship is

confirmed), to Phase III (full-scale, often multicenter, clinical efficacy trials in

hundreds and thousands of patients).

Stage 4: The fourth stage is postapproval. Here the drug is registered and available

on the market. However, even after marketing, the use of the drug is monitored

through formalized pharmacovigilance procedures. Any subsequent clinical

trials (Phase IV) must also comply with GCP.

A brief summary of different stages is shown in Table 1.1.

TABLE 1.1 Stages Defined Within Discovery and Development Programs

Stage I Stage II Stage III Stage IV

Establish discovery

assessment of

compounds with

in vitro and/or

in vivo data

(not regulated

under GxP)

Demonstrate

efficacy, identify

side effects

including Tox and

assessment of

pharmacokinetics

(GLP and GCP)

Gain more data on

safety and

effectiveness in

multicenters with

thousands of

patients (GLP,

GCP, cGMP)

Monitor claims or

demonstrate new

indications;

examine special

drug–drug

interactions; assess

pharmacokinetics

(GLP, GCP, cGMP)

INTRODUCTION 3

Page 26: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

1.1.2 Bioanalytical Laboratories—Bioanalysis

Bioanalytical laboratories have increasingly become center of excellence and

critically important in data generation for discovery, preclinical and clinical devel-

opment in life science industries. Bioanalysis is a broad term that is derived from

analytical applications to biologicalmaterials (matrices) such as human and/or animal

biological fluids and materials (blood, plasma, serum, urine, feces, tissues, etc.),

biopharmaceutical (peptides, protein, etc.), and biochemistry (DNA, RNA, organo-

nucleotides, etc.). The main focus of this book is within the aspects of liquid

chromatography–tandem mass spectrometry (LC–MS/MS) and to certain extent

of immunochemistry assays—enyzme-linked immunosorbent assays (ELISA) or

ligand-binding assays (LBAs). Bioanalysis is mainly referred to the quantitative

determination of drugs and their metabolites, and other life science products in

various sample matrices. However, it should also apply to qualitative analysis

(identification and elucidations) of drug degradants, metabolites, impurities, and

other analytes of interests. The techniques (chromatographic-based and ligand-

binding-based assays) are used very early in the drug discovery and development

process to provide support to product discovery programs on metabolite fate and

pharmacokinetics of chemicals in living cells and animals. They are referred by FDA

Guidance for Industry Bioanalytical Method Validation for chromatographic-based

and ligand-binding-based assays [5]. Their uses continue throughout the nonclinical

and clinical product development phases into postmarketing support and may

sometimes extend into clinical therapeutic monitoring. Recent developments and

industry trends for rapid sample throughput and data generation are introduced and

discussed in following chapters, together with examples of how these high throughput

needs are met in bioanalysis.

1.1.2.1 High-Throughput Bioanalytical Sample Preparation Methods and

automation strategies are authoritative reference on the current state-of-the-art in

sample preparation techniques for bioanalysis. The following related chapters focus

on high-throughput (rapid productivity) techniques and describe exactly how to

perform and automate these methodologies, including useful strategies for method

development and optimization. A thorough review of the literature is included

describing high-throughput sample preparation techniques: protein removal by

precipitation; equilibrium dialysis and ultrafiltration; liquid–liquid extraction; solid

phase extraction; and various online techniques. A schematic diagram of analytical/

bioanalytical techniques used in automation is shown in Figure 1.1.

Among the sample preparation scheme, protein precipitation (PPT) is the most

commonly used approach for a simple, fast, and unique process of removing

unwanted materials from analyte(s) of interest for analysis or in some case for

further cleanup. High selectivity and sensitivity are also imperative for bioanalytical

laboratories to deal with sample analyses with great demand in method limits of

quantitation (LOQ), wide dynamic range (linearity and range), free of interferences

(specificity and selectivity), and other highly challenging requirements such as

multiple compounds (analytes—parent drugs, prodrugs, and their degradants/

4 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

Page 27: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

metabolites), various sample types (matrices), and different analytical techniques

including LC–MS/MS,GC–MS/MS, LC–NMR, ICP–MS, and other advanced hybrid

techniques. In addition to above analytical techniques, immunoassays (ELISA and/or

ligand-binding assays—LBAs or alike) are also widely used within bioanalytical

laboratories, especially in biopharmaceutical and biotechnology industries where

relatively large molecules are dealt such as peptides and proteins as part of

drug development compounds and applying to different therapeutic areas. Rapid

advances in chromatographic as well as ligand-binding assay technologies have

been observed to meet the needs in product research and development processes.

More details of description are elaborated on above analytical and bioanalytical

techniques as powerful methodologies in trace level qualitative and quantitative

analyses.

There have been varieties of separation and detection techniques involved in

analytical and bioanalytical methodologies as indicated in Figure 1.2. More recent

years, biomarker analysis in various therapeutic areas has become incredibly

significant in drug/product development and monitoring programs. Without any

doubt, this has increasingly become part of bioanalytical capabilities. Biomarker

Analytical/Bioanalytical Sample Prep. Chemistry and Techniques

Protein Precipitation Liquid–LiquidExtraction

Solid Phase Extraction

Automation: Liquid Handling Workstations and Robots

TomtecQuadra 96 Plus

Packard MultiProbe IIEx

TecanGenesis Freedom

HamiltonSTAR

FIGURE 1.1 General schematic of analytical/bioanalytical techniques used in laboratory

operations/automation.

Analytical/Bioanalytical Separation/Detection Techniques

Spectroscopy/LBAsSpectrophotometryChromatography

GC, HPLC, CE, UV–VIS, FT-IR, NMR, MS/MS, LBAs, etc.

Small and Large Molecules

Ionic and PolarSpecies

Volatile andNonvolatile

Liquid, Gas, and Solids

FIGURE 1.2 Commonly used techniques in analytical/bioanalytical separation and

detection.

INTRODUCTION 5

Page 28: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

measurements now support key decisions throughout the drug development process,

from lead optimization to regulatory approvals. They are essential for documenting

exposure–response relationships, specificity and potency toward themolecular target,

untoward effects, and therapeutic applications. In a broader sense, biomarkers

constitute the basis of clinical pathology and laboratory medicine. The utility of

biomarkers is limited by their specificity and sensitivity toward the drug or disease

process and by their overall variability. Understanding and controlling sources of

variability is not only imperative for delivering high-quality assay results, but

ultimately for controlling the size and expense of research studies. Variability in

biomarker measurements is affected by biological and environmental factors (e.g.,

gender, age, posture, diet, and biorhythms), sample collection factors (e.g., preser-

vatives, transport and storage conditions, and collection technique), and analytical

factors (e.g., purity of reference material, pipetting precision, and antibody speci-

ficity). The quality standards for biomarker assays used in support of nonclinical

safety studies fall under GLP (FDA) regulations, whereas, those assays used to

support human diagnostics and healthcare are established by Clinical Laboratory

Improvement Amendments (CLIAs) and Centers for Medicare &Medicaid Services

(CMSs) regulations and accrediting organizations such as the College of American

Pathologists (CAPs). While most research applications of biomarkers are not

regulated, biomarker laboratories in all settings are adopting similar laboratory

practices in order to deliver high-quality data. Because of the escalation in demand

for biomarker measurements, the highly parallel (multiplexed) assay platforms that

have fueled the rise of genomics will likely evolve into the analytical engines that

drive the biomarker laboratories of tomorrow. The role of biomarkers in drug

discovery and development has gained precedence over the years. As biomarkers

become integrated into drug development and clinical trials, quality assurance and, in

particular, assay validation become essential with the need to establish standardized

guidelines for bioanalytical methods used in biomarker measurements. New bio-

markers can revolutionize both the development and use of therapeutics but are

contingent on the establishment of a concrete validation process that addresses

technology integration and method validation as well as regulatory pathways for

efficient biomarker development. Perspective focuses on the general principles of the

biomarker validation process with an emphasis on assay validation and the collab-

orative efforts undertaken by various sectors to promote the standardization of this

procedure for efficient biomarker development. It is important to point out that

biomarker method validation is distinct from pharmacokinetic validation and routine

laboratory validation. The FDA has issued guidance for industry [5] on bioanalytical

method validation for assays that support pharmacokinetic studies that are specific for

chromatographic and ligand-binding assays, and that are not directly related to the

qualification or validation of biomarker assays.Whereas routine laboratory validation

refers to laboratories that do testing on human specimens for diagnosis, prevention, or

treatment of any disease and falls under the jurisdiction of the Clinical Laboratory

Improvement Amendments of 1988, there is little regulatory guidance on biomarker

assay validation. Hence, a “fit-for-purpose” approach for biomarker method devel-

opment and validation is derived with the idea that assay qualification or validation

6 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS

Page 29: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

should be tailored to meet the intended purpose of the biomarker study. Numerous

applications using bioanalytical techniques have generated enormous interests and

some case reveal ultimate solutions in drug efficacies and other indications that are

critical to the success in drug/product development and approval processes.

1.1.3 Good Laboratory Practices Versus Bioanalytical Labs/Bioanalysis

Recently, more and more debate and discussion around the connection between GLP

and Bioanalysis are surfaced. It is noted that there is no direct reference from GLP

regulations to bioanalysis. However, it has become common terminology and

acceptance when people refer to GLP–Bioanalysis. In a regulatory term, it may be

referred as regulated bioanalysis to support programs or studies under GLP compli-

ance. There is a misconception in some quarters that GLP is required for the conduct

of clinical studies. This is not correct. The introduction to the OECD Principles of

GLP (and the introduction to the USFDA GLPs in 21 CFR part 58) makes clear that

they apply only to the portions of nonclinical (preclinical) studies. The relevant

documents for clinical studies are the various codes of GC(R)P (e.g., ICH; TGA). The

USFDA and other registration authorities do require a demonstration of the quality of

test data from clinical studies. In the United States, this may well be by means of

conformance with Clinical Laboratories Improvement Act (CLIA) [6]. In Australia,

this is best demonstrated by the testing laboratory’s NATA accreditation (in Medical

Testing, Chemical Testing, etc.)1. Nevertheless, bioanalytical laboratories generate

data in support of clinical studies and ultimately as part of data submissions to

regulatory agencies. More detailed discussions on above techniques and guidelines

are available in respective chapters of this book. The regulatory environment in which

clinical trials are conducted continues to evolve. The changes are generally focused on

requiring more rigorous control within the organizations performing clinical trials in

order to ensure patient safety and the reliability of data produced. The global

acceptance of the ICH Guideline for GCP and the implementation of the European

Union Clinical Trials Directive (2001/20/EC) are two clear examples of such change.

For some years, it has been internationally recognized that clinical laboratories

processing specimens from clinical trials require an appropriate set of standards to

guide good practices. With that aim in mind, the Good Clinical Laboratory Practice

Guidelines [7] were drafted and published in 2003 by a working party of the Clinical

Committee of the British Association of ResearchQuality Assurance (BARQA). This

guidance identifies systems required and procedures to be followed within an

organization conducting analysis of samples from clinical trials in compliance with

the requirements of GCP. It thus provides sponsors, laboratory management, project

managers, clinical research associates (CRAs), and quality assurance personnel with

the framework for a quality system in analysis of clinical trial samples, ensuring GCP

compliance overall of processes and results.

1 The National Association of Testing Authorities (NATA)—Australia’s national laboratory accreditation

authority. NATA accreditation recognizes and promotes facilities competent in specific types of testing,

measurement, inspection, and calibration.

INTRODUCTION 7

Page 30: REGULATED BIOANALYTICAL LABORATORIESdownload.e-bookshelf.de/download/0000/5814/63/L-G-0000581463...2.3 Comparison of FDA GLP, EPA GLP Regulations, and OECD GLP Principles 47 2.3.1

1.2 OBJECTIVES AND KEY REQUIREMENTS FOR GLP

REGULATIONS

The ability to provide timely, accurate, and reliable data is essential to the role of

analytical and bioanalytical chemists and is especially true in the discovery, devel-

opment, and manufacture of pharmaceuticals and life science products. Analytical

and bioanalytical data are used to screen potential drug candidates, aid in the

development of drug syntheses, support formulation studies, animal PK/Tox, clinical

safety and efficacy programs, monitor the stability of bulk pharmaceuticals and

formulated products, and test final products for release. The quality of analytical and

bioanalytical data is a key factor in the success of a drug or product development

program. The process of method development and validation has a direct impact on

the quality of these data.

Although a thorough validation cannot rule out some potential problems, the

process ofmethod development and validation should address themost commonones.

Examples of typical problems that can be minimized or avoided are synthesis

impurities that coelute with the analyte peak in an HPLC assay; a particular type

of column that no longer produces the separation needed because the supplier of the

column has changed themanufacturing process; an assaymethod that is transferred to

a second laboratory where they are unable to achieve the same detection limit; and a

quality assurance audit of a validation report that finds no documentation on how the

method was performed during the validation.

Problems increase as additional people, laboratories, and equipment are used

to perform the method. When the method is used in the developer’s laboratory, a

small adjustment can usually be made to make the method work, but the flexibility

to change it is lost once the method is transferred to other laboratories or used for

official product testing. This is especially true in the pharmaceutical and life

science industries, where methods are submitted to regulatory agencies and changes

may require formal approval before they can be implemented for official testing/

intended use. The best way to minimize method problems is to perform adequate

validation experiments during development and establishment. Analysis of chemi-

cals/drugs in the complex environments/matrices in which they occur are carried out

by a vast range of institutions for a variety of purposes, from pharmaceutical and

agrochemical companies to hospital biochemistry labs and industry laboratories, from

environmental monitoring to safety and toxicity testing of new drugs/products. The

range of compounds for analysis is enormous, from naturally occurring compounds

such as vitamins to man-made chemicals from the pharmaceutical and agrochemical

industries. The following chapters offer an integrated, readable reference text

describing the full range of analytical techniques and regulatory requirements

available for such small molecules (mostly) and large molecules in an up-to-date

manner and should be useful and appeal to all involved in the rapidly growing field of

bioanalytical sciences.

. Responsibilities should be defined for the sponsor management, for the study

management, and for the quality assurance unit.

8 INTRODUCTION, OBJECTIVES, AND KEY REQUIREMENTS FOR GLP REGULATIONS