Atnaujinkite slapukų nuostatas

El. knyga: Early Drug Development: Strategies and Routes to First-in-Human Trials

  • Formatas: PDF+DRM
  • Išleidimo metai: 03-Aug-2010
  • Leidėjas: John Wiley & Sons Inc
  • Kalba: eng
  • ISBN-13: 9780470613177
Kitos knygos pagal šią temą:
  • Formatas: PDF+DRM
  • Išleidimo metai: 03-Aug-2010
  • Leidėjas: John Wiley & Sons Inc
  • Kalba: eng
  • ISBN-13: 9780470613177
Kitos knygos pagal šią temą:

DRM apribojimai

  • Kopijuoti:

    neleidžiama

  • Spausdinti:

    neleidžiama

  • El. knygos naudojimas:

    Skaitmeninių teisių valdymas (DRM)
    Leidykla pateikė šią knygą šifruota forma, o tai reiškia, kad norint ją atrakinti ir perskaityti reikia įdiegti nemokamą programinę įrangą. Norint skaityti šią el. knygą, turite susikurti Adobe ID . Daugiau informacijos  čia. El. knygą galima atsisiųsti į 6 įrenginius (vienas vartotojas su tuo pačiu Adobe ID).

    Reikalinga programinė įranga
    Norint skaityti šią el. knygą mobiliajame įrenginyje (telefone ar planšetiniame kompiuteryje), turite įdiegti šią nemokamą programėlę: PocketBook Reader (iOS / Android)

    Norint skaityti šią el. knygą asmeniniame arba „Mac“ kompiuteryje, Jums reikalinga  Adobe Digital Editions “ (tai nemokama programa, specialiai sukurta el. knygoms. Tai nėra tas pats, kas „Adobe Reader“, kurią tikriausiai jau turite savo kompiuteryje.)

    Negalite skaityti šios el. knygos naudodami „Amazon Kindle“.

Drug development is a highly resource intensive effort. The high attrition rate during nonclinical and especially clinical evaluation demands that the candidate drug selection process he as cost and time effective as possible. Focusing on the critical early stages of drug development, this book guides investigators through the continuum of disciplines that play a role in determining whether a new chemical entity with demonstrated pharmacological activity should progress to clinical evaluation in human subjects. Moreover, it will help them prepare and submit an Investigational New Drug (IND) application.

The book is divided into five parts:

Part I: Introduction

Part II: Lead Optimization Strategies

Part III: Bridging from Discovery to Development

Part IV: Pre-IND Drug Development

Part V: Planning the First-in-Human Study and Regulatory Submission

Each chapter, written by a leading expert, examines a specific discipline, such as ADME and pharmacokinetics, bioanalysis, safey pharmacology, toxicology and toxicokinetics, chemistry/manufacturing and controls (CMC), and exploratory INDs. Authors explore strategies, study designs, challenges, and decision-making criteria for both small molecules and, where relevant, biopharmaceutics, offering specific examples for illustration. They also discuss the interdisciplinary coordination needed to develop a candidate drug that has the best potential of demonstrating a high level of efficacy and safey in its target patient population. In addition, most chapters guide readers through relevant international regulatory guidelines and requirements to help streamline the approval process.

This book is recommended for all investigators involved in nonclinical and clinical drug development, helping them focus their scientific efforts and resources on the best drug candidates and minimize costly attrition during the later stages of drug development.

Mitchell N. Cayen, PhD, is Principal of Cayen Pharmaceutical Consulting, LLC, a firm that advises pharmaceutical companies in nonclinical and clinical drug development, with specific focus on candidate drug selection, discovery, drug metabolism and pharmacokinetics, drug development strategies, and regulatory submissions. Previously, he was senior director of drug metabolism and pharmacokinetics at Schering-Plough Research Institute.

The focus of early drug development has been the submission of an Investigational New Drug application to regulatory agencies. Early Drug Development: Strategies and Routes to First-in-Human Trials guides drug development organizations in preparing and submitting an Investigational New Drug (IND) application. By explaining the nuts and bolts of preclinical development activities and their interplay in effectively identifying successful clinical candidates, the book helps pharmaceutical scientists determine what types of discovery and preclinical research studies are needed in order to support a submission to regulatory agencies.

Recenzijos

"As such, it may serve both as an introduction for newcomers but also as a reference for the more experienced ... The reader will find valuable advice on how to find the best strategy towards entry into the clinic." (ChemMedChem, 2011)

Contributors xix
Foreword xxi
Preface xxiii
PART I INTRODUCTION
1 Drug Discovery and Early Drug Development
3(24)
Mitchell N. Cayen
1.1 The Drug Discovery and Development Scene
3(5)
1.1.1 Pharmaceutical Research and Development Challenges
3(2)
1.1.2 Attrition During Discovery and Development
5(1)
1.1.3 Corporate Strategy Perspectives
6(2)
1.2 Drug Discovery
8(4)
1.2.1 Target Identification
8(1)
1.2.2 Hit-to-Lead Identification
9(1)
1.2.3 Lead Optimization Strategies
10(2)
1.3 Pre-FIH Drug Development
12(3)
1.3.1 Introduction
12(1)
1.3.2 Pre-FIH Toxicology
12(1)
1.3.3 Formulation and Drug Delivery
13(1)
1.3.4 Pre-FIH Drug Metabolism and Pharmacokinetics
14(1)
1.4 The FIH Trial
15(1)
1.5 The Regulatory Landscape
16(2)
1.6 Contract Research Organizations
18(4)
1.7 Concluding Remarks on Introductory Perspectives
22(5)
References
23(4)
PART II LEAD OPTIMIZATION STRATEGIES
2 ADME Strategies in Lead Optimization
27(62)
Amin A. Nomeir
2.1 Introduction
27(3)
2.2 Absorption
30(6)
2.2.1 Permeability
32(3)
2.2.2 Efflux Transport
35(1)
2.3 Distribution
36(6)
2.3.1 Plasma Protein Binding
36(4)
2.3.2 Brain Uptake
40(1)
2.3.3 Tissue Distribution
41(1)
2.4 Metabolism
42(19)
2.4.1 In Vitro Metabolism Studies
42(19)
2.5 Excretion
61(3)
2.6 Pharmacokinetics
64(4)
2.7 Prioritizing ADME Screens
68(1)
2.8 In Silico ADME Screening
69(7)
2.9 The Promise of Metabolomics
76(2)
2.10 Conclusions
78(11)
References
79(10)
3 Prediction of Pharmacokinetics and Drug Safety in Humans
89(42)
Peter L. Bullock
3.1 Introduction
89(2)
3.2 Prediction of Human Pharmacokinetic Behavior
91(22)
3.2.1 In Vitro Models for Predicting Intestinal Absorption, Intrinsic Hepatic Clearance, and Drug Interactions
92(15)
3.2.2 In Vivo Models for Predicting Pharmacokinetic Behavior
107(6)
3.3 Prediction of Drug Safety
113(7)
3.3.1 In Vitro Approaches for Predicting Drug Safety
114(2)
3.3.2 In Vivo and Ex Vivo Methods for Predicting Drug Safety
116(3)
3.3.3 In Silico Methods for Predicting Drug Safety
119(1)
3.4 Conclusions
120(11)
References
121(10)
4 Bioanalytical Strategies
131(76)
Christopher Kemper
4.1 Introduction
131(2)
4.1.1 Bioanalysis: The Primary Basis for Pharmacokinetic and Pharmacodynamic Evaluations
131(1)
4.1.2 Regulatory Initiatives in Bioanalysis
132(1)
4.2 Basic Bioanalytical Techniques and Method Development
133(23)
4.2.1 Sample Preparation
133(6)
4.2.2 Component Separation
139(5)
4.2.3 Detection
144(5)
4.2.4 Ligand-Binding Assays
149(5)
4.2.5 Integration of Method Development Components: Example with LC-MS/MS
154(2)
4.3 Bioanalytical Method Validation
156(12)
4.3.1 Introduction to Validation
156(1)
4.3.2 The Primary Metrics: Acceptance Criteria
157(8)
4.3.3 Additional Validation Criteria
165(3)
4.4 Special Issues with Ligand-Binding Assays
168(1)
4.4.1 Characterization
168(1)
4.4.2 Selectivity Issues
168(1)
4.4.3 Matrix Effects
168(1)
4.4.4 Quantification Issues
169(1)
4.5 Partial and Cross-Validations
169(1)
4.6 Application of Validated Methods to Sample Analyses: Some Perspectives
170(18)
4.6.1 Stability
171(1)
4.6.2 Calibration Curves
172(1)
4.6.3 Quality Control Samples
172(1)
4.6.4 Analytical Notes
172(1)
4.6.5 Acceptance Criteria
173(1)
4.6.6 Repeat Analyses of Incurred Samples
174(2)
4.6.7 Sample Stability and Incurred Samples
176(1)
4.6.8 Scientific Versus Production Issues
177(1)
4.6.9 Documentation
178(1)
4.6.10 Resources
179(9)
4.7 Risk-Based Paradigms: Discovery and Development Support
188(6)
4.7.1 Logistics and Discovery
189(3)
4.7.2 Early Involvement of Consultants and CROs
192(1)
4.7.3 Metabolites: Bioanalytical Issues Pre-FIH
193(1)
4.7.4 Racemic Mixtures
194(1)
4.8 The Road to "First in Human"
194(2)
4.8.1 Clinical Collaboration Prior to Initiation of the FIH Trial
195(1)
4.9 International Perspectives
196(2)
4.9.1 European Union
196(1)
4.9.2 Japan
197(1)
4.9.3 India
197(1)
4.10 Conclusions
198(9)
References
199(8)
PART III BRIDGING FROM DISCOVERY TO DEVELOPMENT
5 Chemistry, Manufacturing, and Controls: The Drug Substance and Formulated Drug Product
207(42)
Orn Almarsson
Christopher J. Galli
5.1 Introduction
207(1)
5.2 Pre-NCE Activities and CMC Development
208(8)
5.2.1 Rationale for CMC Involvement in Discovery
208(1)
5.2.2 Pharmaceutical Properties
209(3)
5.2.3 CMC Interactions with Discovery at NCE Selection
212(2)
5.2.4 Biopharmaceuticals
214(2)
5.3 CMC Considerations at the NCE Stage
216(6)
5.3.1 Solid-State Compounds
216(1)
5.3.2 Selection of Development Form (Crystalline State)
217(3)
5.3.3 Characterization of Drug Substance (Preformulation)
220(2)
5.4 NCE-to-GLP Transition (Bridging from Discovery to Pre-FIH Development)
222(7)
5.4.1 Drug Synthesis and Formulation for Toxicity Studies: Meeting the Delivery Objectives
222(2)
5.4.2 Bridging to Formulations for FIH Studies
224(5)
5.5 CMCs to Meet Clinical Trial Material Requirements
229(7)
5.5.1 Drug Substance Comparability with Material Used in Pre-FIH GLP Studies
229(1)
5.5.2 Good Manufacturing Practices
230(1)
5.5.3 Analytical Development for Assay of Drug Substance and Drug Product
230(5)
5.5.4 Placebos and Blinding
235(1)
5.6 CMC Strategic Considerations
236(2)
5.6.1 Interactions Across Disciplines
236(1)
5.6.2 Outsourcing (and Insourcing) CMC Work
237(1)
5.7 Case Studies
238(6)
5.7.1 Indinavir
238(3)
5.7.2 Doxorubicin Peptide Conjugate
241(3)
5.8 Evolution of Drug Development: Implications for CMCs in the Future
244(5)
Resources
245(2)
References
247(2)
6 Nonclinical Safety Pharmacology Studies Recommended for Support of First-in-Human Clinical Trials
249(34)
Duane B. Lakings
6.1 Introduction and Overview
249(3)
6.2 Timing of Safety Pharmacology Studies
252(2)
6.3 CNS Safety Pharmacology
254(1)
6.4 Cardiovascular Safety Pharmacology
254(13)
6.4.1 Study Designs
254(13)
6.4.2 Additional Information on QT-Interval Prolongation or Delayed Ventricular Repolarization
267(1)
6.5 Respiratory System Safety Pharmacology
267(7)
6.6 Renal/Urinary Safety Pharmacology
274(1)
6.7 Gastrointestinal System Safety Pharmacology
274(1)
6.8 Autonomic Nervous System Safety Pharmacology
275(1)
6.9 Other Systems
276(1)
6.10 Discussion and Conclusions
277(6)
References
279(4)
PART IV PRE-IND DRUG DEVELOPMENT
7 Toxicology Program to Support Initiation of a Clinical Phase I Program for a New Medicine
283(26)
Hugh E. Black
Stephen B. Montgomery
Ronald W. Moch
7.1 Introduction
283(1)
7.2 Toxicology Support of Discovery
284(1)
7.3 Goals of the Pre-FIH Toxicology Program
285(1)
7.4 Importance of a Clinical Review of the Nonclinical Pharmacology Data
286(1)
7.5 Take the Time to Plan Appropriately
286(1)
7.6 The Active Pharmaceutical Ingredient
286(2)
7.6.1 Availability Issues
286(1)
7.6.2 Impurity Considerations
287(1)
7.6.3 Inactive Ingredients
288(1)
7.7 Timely Conduct of In Vitro Assays
288(2)
7.7.1 Comparative In Vitro Metabolism
288(1)
7.7.2 Genetic Toxicology
289(1)
7.8 Development of Validated Bioanalytical and Analytical Assays
290(1)
7.8.1 Validated Bioanalytical Assay for Determining Plasma Concentrations of the NCE
290(1)
7.8.2 Validated Analytical Assays for Dosing Solutions or Suspensions
290(1)
7.8.3 Validated Assays for Dosing Solution Stability
291(1)
7.9 Planning for the Conduct of Toxicity Studies
291(2)
7.9.1 Timing of the IND/CTA
291(1)
7.9.2 The Danger of Shortcuts
292(1)
7.9.3 Pilot In Vivo Studies for Dose Selection and Bleeding Time Determinations
292(1)
7.10 GLP Toxicology Program
293(11)
7.10.1 Toxicology Requirements for Initiating an FIH Trial
294(1)
7.10.2 Toxicology Protocols
295(7)
7.10.3 Study Monitoring
302(1)
7.10.4 Microscopic Examination of Tissues
303(1)
7.10.5 Considerations of the NOAEL and MTD in Protocol Design
303(1)
7.11 Pre-IND Meeting
304(1)
7.12 Conclusions
305(4)
References
306(3)
8 Toxicokinetics in Support of Drug Development
309(52)
Gary Eichenbaum
Vangala Subrahmanyam
Alfred P. Tonelli
8.1 Introduction
309(1)
8.2 Historical Perspectives
310(1)
8.3 Regulatory Considerations
311(1)
8.4 Factors to Consider in the Design of Toxicokinetic Studies
312(20)
8.4.1 Drug Supply Requirements
312(1)
8.4.2 Species Selection
313(1)
8.4.3 API Properties: Salt/Crystal Form, Particle Size, and Impurities
314(1)
8.4.4 Dose-Related Exposure
314(1)
8.4.5 Changes in Pharmacokinetics Following Multiple Dosing
315(1)
8.4.6 Selection of Dosing Vehicles
316(1)
8.4.7 Bioanalytical Method
316(1)
8.4.8 Evaluation of Metabolites
317(4)
8.4.9 Evaluation of Enantiomers
321(1)
8.4.10 Matrix Considerations
321(1)
8.4.11 Number of Animals
322(1)
8.4.12 Gender
322(1)
8.4.13 Dose Selection
323(1)
8.4.14 Dose Volume
324(1)
8.4.15 Blood Sampling Variables
324(5)
8.4.16 Sampling Times
329(2)
8.4.17 Considerations with Biopharmaceutics
331(1)
8.4.18 Practical Considerations in Planning a Toxicokinetic Program
332(1)
8.5 Toxicokinetic Parameter Estimates and Calculations
332(7)
8.5.1 Data Analysis (Noncompartmental Versus Compartmental)
332(1)
8.5.2 Noncompartmental Kinetic Parameters
333(5)
8.5.3 Statistics and Outliers
338(1)
8.5.4 Physiologically Based Toxicokinetic Modeling
338(1)
8.6 Interpretation of Toxicokinetic Data
339(6)
8.6.1 Review of In-life Results
339(1)
8.6.2 Protocol Deviations
339(1)
8.6.3 Confirmation of Exposure and Evaluation of Dose Proportionality
339(2)
8.6.4 Exposure after Single and Multiple Dosing: Accumulation Perspectives
341(2)
8.6.5 Gender Effects
343(1)
8.6.6 Relationship to Toxicology Findings
344(1)
8.6.7 Midstudy Changes in Dosing Duration or Dose Level
345(1)
8.7 Role of Toxicokinetics in Different Types of Toxicity Studies
345(5)
8.7.1 Acute Studies
346(1)
8.7.2 Dose-Range-Finding and Tolerability Studies
346(1)
8.7.3 Subchronic Studies (Two Weeks to Three Months)
347(1)
8.7.4 Chronic Studies (Six to 12 Months)
347(1)
8.7.5 Safety Pharmacology and Specialty Studies
347(1)
8.7.6 Genetic Toxicology
348(1)
8.7.7 Reproductive Toxicology
348(1)
8.7.8 Carcinogenicity Studies
349(1)
8.7.9 Bridging Toxicity Studies
350(1)
8.8 Role of Toxicokinetics in Integrated Safety Assessment
350(5)
8.8.1 Safety Margins: Role in Setting Clinical Doses for FIH Studies
350(2)
8.8.2 Role of Protein Binding and Blood Partitioning
352(1)
8.8.3 Toxicokinetics: Caution about Safety Margins
353(1)
8.8.4 Safety Margins for Different Toxicity Profiles
354(1)
8.9 Conclusions
355(6)
References
355(6)
9 Good Laboratory Practice
361(62)
Anthony B. Jones
Kathryn Hackett-Fields
Shari L. Perlstein
9.1 Introduction
361(2)
9.2 Hazard and Risk
363(3)
9.3 U.S. GLP Regulations
366(21)
9.3.1 Subpart A: General Provisions
367(2)
9.3.2 Subpart B: Organization and Personnel
369(7)
9.3.3 Subpart C: Facilities
376(1)
9.3.4 Subpart D: Equipment
376(1)
9.3.5 Subpart E: Testing Facilities Operation
377(1)
9.3.6 Subpart F: Test and Control Articles
378(1)
9.3.7 Subpart G: Protocol for and Conduct of a Nonclinical Laboratory Study
379(5)
9.3.8 Subpart J: Reports and Records
384(3)
9.3.9 Disqualification of Testing Facilities
387(1)
9.4 GLPs in the Bioanalytical Laboratory
387(6)
9.4.1 Organization and Personnel
389(1)
9.4.2 Equipment and Testing Facilities Operation
389(2)
9.4.3 Some Challenges in the Bioanalytical Laboratory
391(2)
9.5 Moving Into the Future: A Closing Overview
393(2)
9.6 Appendixes
395(28)
Appendix 9.1 Preambles---Perspectives on GLP Requirements
395(1)
Appendix 9.2 International Regulations
396(2)
Appendix 9.3 Paraphrased FDA GLP Definitions
398(1)
Appendix 9.4 FDA Inspections
399(2)
Appendix 9.5 Critical Phase Inspections---What, Why, How, and When?
401(1)
Appendix 9.6 Test System
402(1)
Appendix 9.7 21 CFR Part 11
402(6)
Appendix 9.8 SOP Generation and Review
408(3)
Appendix 9.9 Study Director's Responsibilities
411(2)
Appendix 9.10 Regulatory Requirements for the Study Protocol
413(3)
References
416(7)
PART V PLANNING THE FIRST-IN-HUMAN STUDY AND REGULATORY SUBMISSION
10 Estimation of Human Starting Dose for Phase I Clinical Programs
423(42)
Lorrene A. Buckley
Parag Garhyan
Rafael Ponce
Stanley A. Roberts
10.1 Introduction
423(1)
10.2 Characteristics of Well-Behaved Therapeutic Candidates
424(2)
10.3 Regulatory Guidances for FIH-Enabling Nonclinical Safety Assessment: General Principles
426(1)
10.4 Nonclinical Pharmacokinetics and Pharmacodynamics for Human Dose Projection
427(1)
10.5 Establishing the First-in-Human Dose
427(12)
10.5.1 Phase I Clinical Trial Support: Use of the NOAEL-Based Approach
428(4)
10.5.2 Estimating a Human Dose
432(7)
10.6 Phase I Clinical Trial Support: Use of the MABEL or Pharmacologically Active Dose
439(6)
10.6.1 Predicting the MABEL and PAD in Humans
441(4)
10.7 Support of Exploratory Clinical Studies
445(1)
10.8 Considerations in the Design of Phase I Trials
446(2)
10.8.1 Toxicological Considerations
446(1)
10.8.2 Differences Between Animals and Humans That May Modify Exposure or Response
447(1)
10.8.3 Healthy Human Subjects or Patients
448(1)
10.9 Interdisciplinary Partnerships
448(2)
10.9.1 Chemistry, Manufacturing, and Control
448(1)
10.9.2 Regulatory Affairs
449(1)
10.9.3 Clinical
449(1)
10.10 Beyond the FTH Dose
450(1)
10.11 Concluding Perspective
450(1)
10.12 Four Case Studies
451(14)
References
459(6)
11 Exploratory INDs/CTAs
465(24)
Mitchell N. Cayen
11.1 Introduction
465(2)
11.2 Regulatory Background
467(7)
11.2.1 FDA Single-Dose Toxicity Guidance
467(1)
11.2.2 European Position Paper on Microdose Clinical Trials
467(1)
11.2.3 FDA Critical Path Initiative
468(1)
11.2.4 FDA Guidance on Exploratory IND Studies
469(3)
11.2.5 Belgium National Guidance on Exploratory Trials
472(1)
11.2.6 The ExpIND (or ExpCTA) Submission
473(1)
11.3 Experience and Various Perspectives on ExpINDs or ExpCTAs
474(6)
11.3.1 Microdose Studies
475(4)
11.3.2 Pharmacological Dose and MOA Studies
479(1)
11.4 Some Reactions and Perspectives on the ExpIND/ExpCTA Initiative
480(4)
11.4.1 What an ExpIND/ExpCTA Can Do
481(1)
11.4.2 What an ExpIND/ExpCTA Cannot Do
481(1)
11.4.3 Some Potential Drawbacks or Challenges in the Conduct of an ExpIND/ExpCTA Program
482(2)
11.5 What Is an Ideal Candidate for an ExpIND/ExpCTA?
484(1)
11.6 Conclusions
484(5)
References
486(3)
12 Unique Considerations for Biopharmaceutics
489(24)
Laura P. Andrews
James D. Green
12.1 Introduction and Background
489(1)
12.2 Selection of the Molecule: Contrasts to Small-Molecule Considerations
490(3)
12.2.1 Utility of Animal Efficacy Models
491(1)
12.2.2 In Vitro Activity Profiling, Sequence Homology, and the Use of Homologous Molecules for Nonclinical Efficacy and Safety Assessments
491(1)
12.2.3 In Vivo Profiling of Biopharmaceutical Activity
492(1)
12.3 Production and Process Considerations in Pre-FIH Development
493(2)
12.4 Bioanalytical Assay Considerations
495(1)
12.5 Objectives and Implementation of Pre-FIH Safety Assessment Programs
496(11)
12.5.1 ICH S6 Guideline
496(1)
12.5.2 Considerations and Typical Program Designs for Nonclinical Safety Assessment of Biopharmaceutics
497(10)
12.6 Post-IND Considerations: Support of Phases II and III and Registration
507(1)
12.6.1 Changes in Production and Process, and Impact on Completed Studies
507(1)
12.7 The TeGenero Incident and Implications for Biopharmaceutic Nonclinical Safety Evaluation Programs
508(1)
12.8 Conclusions
509(4)
References
510(3)
13 Project Management and International Regulatory Requirements and Strategies for First-in-Human Trials
513(30)
Carolyn D. Finkle
Judith Atkins
13.1 Introduction: Initiate Product Development with the End in Mind
513(3)
13.2 Importance of Project Management
516(2)
13.3 FDA Input Early and Often
518(1)
13.4 IND Submission in the United States
519(2)
13.5 Global Clinical Trials
521(2)
13.6 Clinical Trial Applications
523(16)
13.6.1 Europe
523(3)
13.6.2 Canada
526(2)
13.6.3 Australia
528(2)
13.6.4 Latin America
530(4)
13.6.5 China
534(1)
13.6.6 India
535(2)
13.6.7 Japan
537(2)
13.7 Conclusions
539(4)
References
539(4)
14 First-in-Human Regulatory Submissions
543(52)
Mary M. Sommer
Mark Ammann
Ulf B. Hillgren
Kathleen J. Kovacs
Keith Wilner
14.1 Introduction
543(1)
14.2 Submission Strategies
544(5)
14.2.1 Regulatory Environment
545(1)
14.2.2 Clinical Considerations
546(3)
14.3 First-in-Human Dossiers
549(10)
14.3.1 Introduction
549(1)
14.3.2 General Considerations for Dossier Preparations
549(4)
14.3.3 Coordination of the Disciplines
553(4)
14.3.4 Document Preparation
557(2)
14.4 United States: Investigational New Drug Application
559(24)
14.4.1 Regulatory Perspective
559(6)
14.4.2 Chemistry, Manufacturing, and Controls
565(9)
14.4.3 Nonclinical Sections
574(6)
14.4.4 Clinical Components
580(3)
14.5 European Union: Clinical Trial Application
583(5)
14.5.1 Regulatory Perspective
583(3)
14.5.2 Quality
586(1)
14.5.3 Nonclinical Sections
587(1)
14.6 Japan: Clinical Trial Protocol Notification
588(1)
14.6.1 Regulatory Perspective
588(1)
14.6.2 Quality
588(1)
14.6.3 Nonclinical Sections
588(1)
14.7 Emerging Regions
589(1)
14.8 Biopharmaceuticals
589(2)
14.9 Final Considerations
591(4)
14.9.1 Gap Analysis
591(1)
14.9.2 Preparation for Regulatory Queries
592(3)
Appendix 1 Abbreviations and Acronyms 595(6)
Appendix 2 Definitions and Glossary of Terms 601(6)
Appendix 3 Some Relevant Government and Regulatory Documents 607(6)
Appendix 4 Some Relevant Resources with Web Sites 613(4)
Index 617
MITCHELL N. CAYEN, PhD, is Principal of Cayen Pharmaceutical Consulting, LLC, a firm that advises pharmaceutical companies in nonclinical and clinical drug develop-ment, with specific focus on candidate drug selection, discovery, drug metabolism and pharmacokinetics, drug development strategies, and regulatory submissions. Previously, he was senior director of drug metabolism and pharmacokinetics at Schering-Plough Research Institute.