did-you-know? rent-now

Amazon no longer offers textbook rentals. We do!

did-you-know? rent-now

Amazon no longer offers textbook rentals. We do!

We're the #1 textbook rental company. Let us show you why.

9781119755852

Drug Safety Evaluation

by ;
  • ISBN13:

    9781119755852

  • ISBN10:

    1119755859

  • Edition: 4th
  • Format: Hardcover
  • Copyright: 2023-01-12
  • Publisher: Wiley
  • Purchase Benefits
  • Free Shipping Icon Free Shipping On Orders Over $35!
    Your order must be $35 or more to qualify for free economy shipping. Bulk sales, PO's, Marketplace items, eBooks and apparel do not qualify for this offer.
  • eCampus.com Logo Get Rewarded for Ordering Your Textbooks! Enroll Now
List Price: $367.99 Save up to $0.84
  • Buy New
    $367.15
    Add to Cart Free Shipping Icon Free Shipping

    PRINT ON DEMAND: 2-4 WEEKS. THIS ITEM CANNOT BE CANCELLED OR RETURNED.

Supplemental Materials

What is included with this book?

Summary

Drug Safety Evluation

Comprehensive and practical guide presenting a roadmap for safety assessment as an integral part of the development of drugs and therapeutics

This fourth edition of Drug Safety Evaluation maintains the central objective of presenting an all-inclusive practical guide for those who are responsible for ensuring the safety of drugs and biologics to patients, healthcare providers, those involved in the manufacture of medicinal products, and all those who need to understand how the safety of these products is evaluated and shepherding valuable candidates to market.

Individual chapters address specific approaches to evaluation hazards, including problems that are encountered and their solutions. Also covered are the scientific and philosophical bases for evaluation of specific concerns (e.g., carcinogenicity, development toxicity, etc.) to provide both understanding and guidance for approaching the new problems that have come to face both our society and the new challenges they brought.

The many changes in regulatory requirements, pharmaceutical development, technology, and the effects of Covid on our society and science have required both extensive revision to every chapter and the addition of four new chapters.

Specific sample topics covered in Drug Safety Evaluation include:

  • The drug development process and the global pharmaceutical marketplace and regulation of human pharmaceutical safety
  • Sources of information for consideration in study and program design and in safety evaluation
  • Electronic records, reporting and submission, screens in safety and hazard assessment, and formulations, routes, and dosage regimens
  • Mechanisms and endpoints of drug toxicity, pilot toxicity testing in drug safety evaluation, and repeat dose toxicity
  • Genotoxicity, QSAR tools for drug safety, toxicogenomics, nonrodent animal studies, and developmental and reproductive toxicity testing
  • An appendix which provides an up to date guide to CROs for conducting studies

Drug Safety Evaluation was written specifically for the pharmaceutical and biotechnology industries, including scientists, consultants, and academics, to show a utilitarian yet scientifically valid path to the everyday challenges of safety evaluation and the problem solving that is required in drug discovery and development.

Author Biography

Shayne Cox Gad, PhD, DABT is the Principal of Gad Consulting Services. He has more than 47 years of experience as a toxicologist, statistical consultant, manager, and consultant on research and development in the chemical, consumer product, contract testing, biotechnology, medical device, and pharmaceutical industries. He has successfully file 138 INDs and authored and edited 52 books, as well as numerous papers, presentations, and other publications.

Dexter W. Sullivan, Jr., MS, DABT is Senior Toxicologist at Gad Consulting Services.

Table of Contents

PREFACE              xxv

ABOUT THE AUTHOR      xxvii

Chapter 1:           The Drug Development Process and the Global Pharmaceutical Marketplace

1.1          Introduction

1.2          The Marketplace             

1.3          History of Modern Therapeutics

1.4          The Drug Development Process

1.5          Strategies for Development: Large vs. Small Company or the Short vs. Long Game

1.5.1      Do Only What You Must (the short game)

1.5.2      Minimize the Risk of Subsequent Failure

1.6          Safety Assessment and the Evolution of Drug Safety

1.7          The Three Stages of Drug Safety Evaluation in the General Case

 

Chapter 2:           Regulation of Human Pharmaceutical Safety: Routes to Human Use and Market

                2.1 Introduction 

                2.2 Brief History of US Pharmaceutical Law

                                2.2.1 1906: Pure Food and Drug Act

                                2.2.2 1938: Food, Drug, and Cosmetic Act

                                2.2.3 1962: Major Amendment

2.2.4 1992, 1997, 2002, 2007, 2012 and 2017: PDUFA, FDAMA, and FDARA

2.2.5 PREA: the Pediatric Research Equity Act

2.2.6 ICH: the International Conference on Harmonization

2.2.7 Electronic Recordings: Electronic Submission Impact

2.2.8 COVID-19

2.3 FDAMA Summary: Consequences and Other Regulations

                2.4 Overview of US Regulations

                                2.4.1 Regulations: General Considerations

                                2.4.2 Regulations: Human Pharmaceuticals

                                2.4.3 Regulations: Environmental Impact

                                2.4.4 Regulations: Antibiotics

                                2.4.5 Regulations: Biologics

                                2.4.6 Regulations vs. Law

                2.5 Organizations Regulating Drug and Device Safety in the U.S.

                2.6 Process of Pharmaceutical Product Development and Approval

2.7 Testing Guidelines

                                2.7.1 Toxicity Testing: Traditional Pharmaceuticals

                                2.7.2 General or Systematic Toxicity Assessment

                                2.7.3 Genetic Toxicity Assessment

                                2.7.4 Safety Pharmacology

                                2.7.5 Local Tissue Tolerance

2.7.6 Reproductive and Developmental

2.7.7 Carcinogenicity

                                2.7.8 Toxicity Testing: Biotechnology Product

                                2.7.9 Special Cases

                2.8 Toxicity/Safety Testing: Cellular and Gene Therapy Products

                                2.8.1 Cellular Therapies

                                2.8.2 Gene Therapies

                                2.8.3 Ex Vivo

                                2.8.4 In Vivo

                                2.8.5 Preclinical Safety Evaluation

2.8.6 Basic Principles for Preclinical Safety Evaluation of Cellular and Gene Therapies

2.8.7 Additional Considerations for Cellular Therapies

2.8.8 Additional Considerations for Gene Therapieces

                2.9 Toxicity Testing: Special Cases

                                2.9.1 Oral Contraceptives

                                2.9.2 Life-Threatening Diseases (Compassionate Use)

                                2.9.3 Vaccines

                                2.9.4 Oncology Drugs and Imaging Agents

                                2.9.5 Optical Isomers

                                2.9.6 Special Populations: Pediatric and Geriatric Claims

2.9.7 Orphan Drugs

                2.9.8 Expedited and Augmented Routes to Approval

2.9.9 Botanical Drug Products

                2.9.10 Types of New Drug Applications (NDAs)

2.10 International Pharmaceutical Regulation and Registration

                2.10.1 International Conference on Harmonization

                                2.10.1.1 Carcinogenicity Studies

                                2.10.1.2 Chronic Toxicity

                                2.10.1.3 Developmental and Reproductive Toxicity

                2.10.2 Other International Considerations

                                2.10.2.1 European Union

                                2.10.2.2 Japan

                                2.10.2.3 China

                2.10.3 Safety Pharmacology

2.11 Combination Products

2.11.1 Device Programs that CDER and CBRH each will Administer                           

2.11.2 Coordination

2.11.3 Submissions

                2.11.3.1 Center Jurisdiction

                2.11.3.2 General Criteria Affecting Drug/Device Determination

                2.12 Meetings and submissions to FDA for Toxicologists

2.13 Conclusions

 

Chapter 3:           Data Mining: Sources of Information for Consideration in Study and Program Design and in Safety Evaluation

3.1 Introduction

                3.1.1 Claims

                3.1.2 Time and Economies

                                                3.1.3 Prior Knowledge

                                                3.1.4 Miscellaneous Reference Sources

                3.1.5 Search Procedure

3.1.6 Monitoring Published Literature and Other Research in Progress

                3.1.7 Kinds of information

                3.1.8 Toxic Release Inventory (TRI)

                3.1.9 Material Safety Data Sheets (MSDS)

3.1.10 Canadian Centre for Occupational health and Safety (CCINFO)

                3.1.11 Pollution and Toxicology (POLTOX)

                3.1.12 MEDLINE and PubChem

3.2 PC-Based Information Products: Laser DISC

                                                3.2.1 International Veterinary Pathology Slide Bank (IVPSB)

3.3 Conclusions

 

Chapter 4:           Electronic Records, Reporting and Submission: eCTD and SEND

4.1 Introduction

4.2 Submission of SEND data in Module 4 of the eCTD

4.3 SEND Background

4.4 SEND Regulatory

4.5 SEND Features

4.6 SEND Study Submission Package

4.7 Determination of Studies that Need Data to be Submitted as SEND Files

                4.7.1 FDA Center

                4.7.2 Type of Application

                4.7.3 Study Start Date

4.8 Storage of Files at the FDA

4.9 Recommended Regulatory Resources

 

Chapter 5: Screens in safety and hazard assessment

5.1 introduction

5.2 characteristics of screens

5.3 uses of screens

5.4 types of screens

                5.4.1 Single stage

                5.4.2 Sequential

                5.4.3 Tier (or multistage)

5.5 Criterion: Development and Use

5.6 Analysis of Screening Data

5.7 univariate data

                5.7.1 control charts

                5.7.2 central tendency plots

                5.7.3 multivariate data

                5.7.4 the analog plot

Chapter 6:           Formulations, Routes, and Dosage Regimens

6.1 Introduction

6.2 Mechanisms

                6.2.1 Local Effects

                6.2.2 Absorption and Distribution

                                6.2.3 Metabolism

                6.3 Common Routes      

                6.3.1 Dermal Route        

                6.3.2 Parenteral Route

                                6.3.2.1 Intravenous Route

                6.3.3 Bolus vs. Infusion

                                6.3.3.1 Subcutaneous Route

                                6.3.3.2 Intramuscular Route

                                6.3.3.3 Intraperitoneal Route

                6.3.4 Oral Route

                                6.3.4.1 Mechanisms of Absorption

                                6.3.4.2 Factors Affecting Absorption

                                6.3.4.3 Bioavailability and Thresholds

                                6.3.4.4 Techniques of Oral Administration            

                6.3.5 Minor Routes        

                                6.3.5.1 Periocular Route

                                6.3.5.2 Rectal Administration

                                6.3.5.3 Vaginal Administration

                                6.3.5.4 Nasal Administration

                                6.3.5.5 Volume Limitations by Route

                6.3.6 Route Comparison and Contrasts

6.3.6.1 Vehicles that Can Mask the Effects of Active Ingredients

6.4 Formulation of Test Materials

                6.4.1 Preformulation

                6.4.2 Dermal Formulations

                6.4.3 Interactions between Skin, Vehicle and Test Chemical

                6.4.4 Oral Formulations

                6.4.5 Parenteral Formulations

6.5 Dosing Calculations

                6.6 Calculating Material Requirements

6.7 Excipients

                6.7.1 Regulation of Excipients

Chapter 7:           Mechanisms and Endpoints of Drug Toxicity

7.1 Manifestations

7.2 Mechanisms of Toxicity

7.3 End Points Measured in General Toxicity Studies

7.3.1 Clinical Observations

                7.3.2 Body Weights

                                7.3.3 Food and Water Consumption

                                7.3.4 Clinical Signs

                7.3.5 Clinical Chemistry and Pathology

                7.3.6 Hematology                           

                7.3.7 Gross Necropsy and Organ Weights

                7.3.8 Histopathology

                7.3.9 Ophthalmology

                7.3.10 Cardiovascular Function

                7.3.11 Neurotoxicology

                7.3.12 Immunotoxicology

                7.3.13 Imaging and Telemetry

7.4 Complications

 

Chapter 8:           Pilot Toxicity Testing in Drug Safety Evaluation: MTD and DRF

8.1 Introduction

8.2 Range-Finding Studies           

                8.2.1 Lethality Testing

                                8.2.1.1 Classical LD50

                                8.2.1.2 Dose Probes

                                8.2.1.3 Up/Down Method

                                8.2.1.4 “Pyramiding” Studies

                                8.2.1.5 Limit Tests

                                8.2.1.6 Fixed-Dose Procedure

                                8.2.1.7 “Rolling” Acute Test

8.2.2 Using Range-Finding Lethality Data in Drug Development: The Minimum Lethal Dose

                8.2.2.1 Minimum Lethal Dose Protocols

8.3 Acute Systemic Toxicity Characterization

                8.3.1 Minimal Acute Toxicity Test

                                8.3.1.1 Clinical Signs

                8.3.2 Complete Acute Toxicity Testing

                                8.3.2.1 Body Weight Considerations

                                8.3.2.2 Pathology Considerations

                                8.3.2.3 Supplemented Acute Studies

                8.3.3 Acute Toxicity Testing with Nonrodent Species

                8.3.4 Factors that Can Affect Acute tests

                                8.3.4.1 Number, Size, and Sex of Dosage Groups

                8.3.5 Selection of Dosages

                                8.3.5.1 Timing

8.4 Screens

                8.4.1 General Toxicity Screens

                8.4.2 Specific Toxicity Screens

8.5 Pilot and DRF Studies

 

 

Chapter 9:           Repeat Dose Toxicity Studies

9.1 Objectives

9.2 Regulatory Considerations

                9.2.1 Good Laboratory Practices

                9.2.2 Animal Welfare Act             

                9.2.3 Regulatory Requirements for Study Design

                9.3 Study Design and Conduct

                9.3.1 Animals

                9.3.2 Routes and Setting Doses

                9.3.3 Parameters to Measure

                                9.3.3.1 Pharmacokinetics and Metabolism

                9.3.4 Study Designs        

9.4 Study Interpretation and Reporting

                9.5 Read Across for Program Wide Evaluation

 

Chapter 10:         Genotoxicity

                10.1 ICH Test Profile

                10.2 DNA Structure

                10.2.1 Transcription

                10.2.2 Translation

                                10.2.3 Gene Regulation

                                10.2.4 DNA Repair

                                                10.2.4.1 Excision Repair

                10.2.5 Error-Prone Repair

                10.2.6 Mismatch Repair

                10.2.7 The Adaptive Repair Pathway

                10.2.8 Plasmids

                10.2.9 Plasmids and DNA Repair

                10.2.10 Nature of Point Mutations

                10.2.11 Suppressor Mutations

                10.2.12 Adduct Formation

                10.2.13 Mutations Due to Insertion Sequences

                10.2.14 The Link Between Mutation and Cancer

10.2.15 Genotoxic vs. Nongenotoxic Mechanisms of Carcinogenesis

10.2.16 Genetic Damage and Heritable Defects

10.2.17 Reproductive Effects

                10.3 Cytogenetics

                                10.3.1 Cytogenetic Damage and its Consequences

                                10.3.2 Individual Chromosomal Damage

                                10.3.3 Chromosome Set Damage

                                10.3.4 Test Systems

                                10.3.5 In Vitro Test Systems

                                                10.3.5.1 In Vitro Metabolic Activation

                                10.3.6 Bacterial Mutation Tests

                                                10.3.6.1 Reversion Test: Background

                                                10.3.6.2 Genetic Makeup of Tester Strains

                                                10.3.6.3 The Use of the Plasmid pKM101

                                                10.3.6.4 Ames Salmonella/Plate Incorporation Method

                                10.3.7 Controls

                                                10.3.7.1 Positive Controls

                                                10.3.7.2 Untreated/Vehicle Controls

                                                10.3.7.3 Evaluation of Results

                                                10.3.7.4 Preincubation tests

                                                10.3.7.5 E. Coli Tester Strains

                                                10.3.7.6 Storage and Checking of Tester Strains

                                10.3.8 Plate Incorporation Assay

                                                10.3.8.1 Protocol for Dose Ranging and Selection

                                                10.3.8.2 Eukaryotic Mutation Tests

                                10.3.9 Eukaryotic Mutation Tests

10.3.10 In Vitro Tests for the Detection of Mammalian Mutation

                10.3.10.1 Chinese Hamster Lines

                10.3.10.2 V79 System

                10.3.10.3 Preliminary Cytotoxicity Testing

                10.3.10.4 Data Analysis

10.3.10.5 Chinese Hamster Ovary (CHO)/Hypoxanthine-Guanine Phosphoribosyltransferase (HGPRT) Assay

10.3.10.6 Mouse Lymphoma L5178Y TK+/- Assay

10.3.10.7 Selection of Dose Levels

10.3.10.8 Main Mutation Assay

10.3.10.9 In Vivo Genotoxicity Tests for the Assessment of Primary DNA Lesions

10.3.10.10 The Comet Assay

10.3.10.11 Principle of Method

10.3.10.12 Status of Mammalian Mutation Tests

                                10.3.11 In Vivo Mammalian Mutation Tests

                                                10.3.11.1 The Mouse Specific Locus Test

                10.4 In Vitro Cytogenetic Assays

                                10.4.1 Cell Types

                                10.4.2 Chinese Hamster Cell Lines

                                10.4.3 Human Peripheral Blood Lymphocytes

                                10.4.4 Positive and Negative Controls

                                10.4.5 Treatment of Cells

                                10.4.6 Scoring Procedures

                                10.4.7 Data Recording

                                10.4.8 Presentation of Results

                10.5 In Vivo Cytogenetic Assays

                                10.5.1 Somatic Cell Assays

                                                10.5.1.1 Metaphase Analysis

                                                10.5.1.2 Micronuclei

                                10.5.2 Germ Cell Assays

                                10.5.3 Heritable Chromosome Assays

                                10.5.4 Germ Cell Cytogenetic Assays

                10.6 Sister Chromatid Exchange Assays

                                10.6.1 Relevance of SCE in Terms of Genotoxicity

                                10.6.2 Experimental Design

                10.7 How to Deal with Positive Test Results

 

Chapter 11:         QSAR Tools for Drug Safety

11.1 Structure- Activity Relationships

                11.1.1 Basic Assumptions

                                11.1.2 Molecular Parameters of Interest

11.2 SAR Modeling Methods

11.3 Applications in Toxicology

                11.3.1 Metabolism

                11.3.2 Reproductive

                11.3.3 Eye Irritation

                11.3.4 Lethality

                                11.3.4.1 Oral Rat LD50

                11.3.5 Carcinogenicity

11.4 Genotoxicity

                11.4.1 QSAR for Mutagenicity

                                11.4.1.1 Sensitization

                                11.4.1.2 Hepatotoxicity

                                11.4.1.3 Cardiotoxicity

11.5 Comparison of Available Models/Applications

                11.5.1 QSAR of Metabolism        

                                11.5.2 Meteor

                                11.5.3 Derek

                                11.5.4 Leadscope

                                                11.5.4.1 Multiple Computer-Automated Structural Evaluation

                                                11.5.4.2 Toxicity Prediction by Computer-Assisted Technology

                                11.5.5 VEGA

                                                11.5.5.1 Global AD Index

11.5.5.2 Similar Molecules with Known Experimental Value

11.5.5.3 Accuracy of Prediction for Similar Molecules

11.5.5.4 Concordance for Similar Molecules

11.5.5.5 Atom-Centered Fragments Similarity Check

11.5.5.6 Model Descriptors Range Check

                                11.5.6 Derek vs. Leadscope

11.6 Near Neighbor Surrogates and their Use

 

Chapter 12:         Toxicogenomics

                12.1 Introduction

                12.2 Uses of Toxicogenomics

 

Chapter 13:         Immunotoxicology in Drug Development

                13.1 Introduction

                13.2 Overview of the Immune System

                13.3 Immunotoxic Effects

                13.4 Immunosuppression

                13.4.1 Immunosuppressive Drugs

                                13.4.1.1 Antimetabolites

                                13.4.1.2 Glucocorticosteroids

                                13.4.1.3 Cyclosporine

                                13.4.1.4 Nitrogen Mustards

                                13.4.1.5 Estrogens

                                13.4.1.6 Heavy Metals

                                13.4.1.7 Antibiotics

13.5 Immunostimulation

                                13.5.1 Hypersensitivity (or Allergenicity)

                                                13.5.1.1 Type I Hypersensitivity

                                                13.5.1.2 Type II Hypersensitivity

                                                13.5.1.3 Type III Hypersensitivity

                                                13.5.1.4 Type IV Delayed-Type Hypersensitivity (DTH)

                13.5.2 Photosensitization

                13.5.3 Autoimmunity

13.6 Regulatory Positions

                13.6.1 CDER Guidance for Investigational New Drugs

                13.7 Evaluation of the Immune System

                13.7.1 Immunopathologic Assessments

                                13.7.1.1 Organ and Body Weights

13.7.2 Humoral (Innate) Immune Response and Possible Entry Points for Immunotoxic Actions

                13.7.2.1 Hematology

                13.7.2.2 Clinical Chemistry

                13.7.2.3 Histopathology

                13.7.2.4 Antibody Plaque-Forming Cell (PFC) Assay

                13.7.2.5 B-Cell Lymphoproliferation Response

                13.7.3 Cell-Mediated Immunity

                                13.7.3.1 T-Cell Lymphoproliferation Response

                                13.7.3.2 Mixed Lymphocyte Response (MLR) Assay

                                13.7.3.3 Cytotoxic T Lymphocyte (CTL)-Mediated Assay

                                13.7.3.4 Delayed-Type Hypersensitivity (DTH) Response

13.8 Nonspecific Immunity Function Assay

13.8.1 Natural Killer Cell Assays

                13.8.2 Macrophage Function

                13.8.3 Mast Cell/Basophil Function

                                13.8.3.1 Host-Resistance Assays

13.9 T-Cell-Dependent Antibody Response (TDAR)

                13.9.1 Treatment

                13.9.2 Hypersensitivity

                                13.9.2.1 Type I Hypersensitivity

                                13.9.2.2 Types II and III Hypersensitivity

                                13.9.2.3 Type IV Hypersensitivity

                                13.9.2.4 Modified Buehler

                                13.9.2.5 Guinea Pig Maximization Test

                13.9.3 Local Lymph Node Assay (LLNA)

                13.9.4 Photosensitization

                                13.9.4.1 Harber and Shalita Method

                                13.9.4.2 Armstrong Method

13.10 Approaches to Compound Evaluation

                13.10.1 Use of In Vivo Tests

                                13.10.1.1 Species Selection

                                13.10.1.2 Route and Treatment Regimen

                13.10.2 Use of In Vitro Tests

13.10.3 Assessment of Immunotoxicity and Immunogenicity/Allergenicity of Biotechnology-Derived Drugs

13.10.4 Suggested Approaches to Evaluation of Results

                13.11 Problems and Future Directions

                                13.11.1 Data Interpretation

                                13.11.2 Appropriate Animal Models

                                13.11.3 Indirect Immunotoxic Effects

                                13.11.4 Hypersensitivity Tests

                                13.11.5 Anaphylaxis Tests

                                13.11.6 Autoimmunity

                                13.11.7 Functional Reserve Capacity

                                13.11.8 Significance of Minor Perturbations

                                13.11.9 Biotechnology Products and antigenicity

                13.12 Summary

 

Chapter 14:         Nonrodent Animal Studies

14.1 Introduction

14.2 Comparison Between Rodent and Nonrodent Experimental Design

                14.2.1 Number of Animals

                14.3 Differences in Study Activities

                14.3.1 Blood Collection

                14.3.2 Dosing

                14.3.3 Handling of Animals

                14.3.4 Behavioral Evaluation

14.4 Nonrodent Models (Species Selection)

14.5 Dog             

                14.5.1 Environmental and Dietary Requirements

                14.5.2 Common Study Protocols

                14.5.3 General Study Activities

                                14.5.3.1 Dosing Techniques

                                14.5.3.2 Clinical Observations and Physical Examinations

                                14.5.3.3 Feed consumption

                                14.5.3.4 Electrocardiograms

                                14.5.3.5 Blood and Urine Collection

                14.5.4 Advantages and Disadvantages

                14.6 The Ferret

                14.6.1 Environmental and Dietary Requirements

                14.6.2 Study Protocols

                14.6.3 General Study Activities

                                14.6.3.1 Dosing techniques

14.6.3.2 Clinical Observations and Physical Examinations

                                14.6.3.3 Feed consumption

                                14.6.3.4 Electrocardiograms

                                14.6.3.5 Blood and Urine Collection

                14.6.4 Advantages and Disadvantages

                14.7 The Pig

                14.7.1 Background

                                14.7.7.7 Restraint and Dosing

                14.7.2 Clinical Laboratory            

                14.7.3 Xenobiotic Metabolism

                14.7.4 Dermal Toxicity

                14.7.5 Cardiovascular Toxicity

                14.7.6 Advantages and Disadvantages

                14.8 The Rabbit

14.8.1 Husbandry

                14.8.1.1 Facilities

                14.8.1.2 Feed and Water

                14.8.1.3 Handling and Restraint

14.8.2 Dosing techniques

14.8.3 Collection Techniques

14.8.4 Study Designs

14.9 Nonhuman Primates

                14.9.1 Environmental and Dietary Requirements

                14.9.2 Common Study Protocols

                14.9.3 General Study Activities

                                14.9.3.1 Common Dosing Techniques

                                14.9.3.2 Clinical Observations and Examinations

                                14.9.3.3 Feed Consumption

                                14.9.3.4 ECGs and Cardiovascular Measurements

                                14.9.3.5 Blood and Urine Collections

                14.9.4 Advantages and Disadvantages

                14.10 Issues in Animal Model Selection

14.11 Statistics in Large Animal Studies

14.11.1 Reasons for Small Sample Sizes in Large Animal Toxicology

                14.11.2 Cross-Sectional or Longitudinal Analysis?

                14.11.3 Repeated Measures: Advantages

                14.11.4 Repeated Measures: Disadvantages

14.11.5 Common Practices in Large Animal Toxicology

14.11.6 Univariate (Repeated Measures) Techniques: Advantages

14.11.7 Univariate (Repeated Measures) Techniques: Disadvantages

14.11.8 Multivariate Techniques: Advantages

14.11.9 Multivariate Techniques: Disadvantages

14.11.10 Some other design factors to be considered in analysis

14.11.11 Covariates: Advantages

14.11.12 Covariates: Disadvantages

14.11.13 Missing Values

                14.12 Read-across for Data Integration

14.13 Summary

 

Chapter 15:         Developmental and Reproductive Toxicity Testing

15.1 Introduction

15.2 ICH Study Designs

15.2.1 Male and Female Fertility and Early Embryonic Development to Implantation

15.2.2 Embryo-fetal Development

15.2.3 Adverse Effects

15.2.4 Pre- and Postnatal Development

15.2.5 Single-Study and two-Study Designs for Rodents

15.2.6 Preliminary Studies

15.2.7 Potential Male Mediated Developmental Effects

15.2.8 Toxicokinetics

15.2.9 Timing of Studies

                15.3 Methodological Issues

                                15.3.1 Control of Bias

                                15.3.2 Diet

                                15.3.3 Clinical Pathology

                                15.3.4 Gravid Uterine Weights

15.3.5 Implant Counts and Determination of Pregnancy

15.3.6 Fetal Examinations

                15.3.6.1 Examination of External Genitalia

                15.3.6.2 Visceral Fetal Examinations

                15.3.6.3 Skeletal Fetal Examination

15.3.7 Developmental Signs

15.3.8 Behavioral Tests

15.3.9 Detecting Effects on Male Reproduction

15.4 Developmental Studies in Primates

15.5 Data Interpretation

15.5.1 Use of Statistical Analyses

15.5.2 Potential Hazard Categories of Developmental Toxins

15.5.3 Associations between developmental and Maternal Toxicity

15.5.4 Assessment of Human Risk

15.6 Juvenile and Pediatric Toxicology

15.7 In Vitro Tests for Developmental Toxicity

15.8 Appraisal of Current Approaches for Determining Developmental and Reproductive Hazards.

 

Chapter 16:         Carcinogenicity Studies

 16.1 Introduction

                16.1.1 History of Xenobiotic Carcinogenesis

                16.2 Mechanisms and Classes of Carcinogens

16.3 Genotoxic Carcinogens

16.4 Epigenetic Carcinogens

                16.5 Regulatory Requirements and timing

                16.5.1 Waivers of Required Testing

16.6 Species and strain

                16.7 Animal Husbandry

                16.8 Dose

                16.8.1 Number of Dose Levels

                16.8.2 Number of Control Groups            

                16.8.3 Criteria for Dose Selection

                16.9 Group Size

                16.10 Route of Administration

                16.11 Study Duration

                16.12 Survival

                16.13 End Points Measured

                16.14 Transgenic Mouse Models

                                16.14.1 The Tg.AC Mouse Model

                                16.14.2 The Tg.rasH2 Mouse Model

                                16.14.3 The P53+/- Mouse Model

                                16.14.4 The XPA-/- Mouse Model

                16.15 Interpretation of Results: Criteria for a Positive Result

                16.16 Statistical Analysis

                                16.16.1 Exact Tests

                                16.16.2 Trend Tests

                                16.16.3 Life Table and Survival Analysis

                                16.16.4 Peto Analysis

                                                16.16.4.1 Interval Selection for Occult (Internal Organ) Tumors

                                                16.16.4.2 Logistic Regression Method for Occult (Internal organ) Tumors

                                16.16.5 Methods to be Avoided

                                16.16.6 Use of Historical Controls

                                16.16.7 Relevance to Human Controls    

16.17 Weight-of-Evidence Factors for Consideration in a Carcinogenicity Assessment Document (CAD)

16.18 Conclusions

 

Chapter 17:         Histopathology and Clinical Pathology in Nonclinical Pharmaceutical Safety Assessment '

17.1 Introduction

                17.1.1 Pathological Techniques

                                17.1.2 Organ Weights

                17.2 Clinical Pathology

                                17.2.1 Clinical Chemistry

                17.2.2 Target Organ Toxicity Biomarkers

                17.2.3 Integrated Analysis of Available Data

 

Chapter 18:         Irritation and Local Tissue Tolerance in Pharmaceutical Safety Assessment

18.1 Introduction

                18.2 Factors Affecting Irritation Responses and Test Outcome

18.3 Primary Dermal Irritation (PDI) Test

18.4 Other Nonparenteral Route Irritation Tests

18.5 Ocular Irritation Testing

18.6 Vaginal Irritation

18.7 Acute Primary Vaginal Irritation Study in the Female Rabbit

18.7.1 Repeated-dose Vaginal Irritation in the Female Rabbit

18 .7.2 Repeated-dose Vaginal Irritation in the Ovariectomized Rats

                18.8 Parenteral Irritation/Tolerance

                                18.8.1 Parenteral Routes

                                                18.8.1.1 Irritation

                                                18.8.1.2 Blood Compatibility

                                                18.8,1.3 Sterility

                                18.8.2 Test Systems for Parenteral Irritation                        

                                                18.8.2.1 Acute Intramuscular Irritation in the Male Rabbit

                                                18.8.2.2 Acute Intravenous Irritation in the Male Rabbit

                18.9 Problems in Testing (and Their Resolutions)

                                18.9.1 Alternative to In Vivo Parenteral Tests

                18.10 Phototoxicity

                                18.10.1Theory and Mechanisms

18.10.2 Factors Influencing Phototoxicity/Photosensitization

                18.10.3 Predictive Tests for Phototoxicity

                18.10.4 3/t3 In Vitro Test

                18.10.5 Rabbit Phototoxicity Test

                18.10.6 Guinea Pig

                18.10.7 Pyrogenicity

                                18.10.7.1 Apparatus and Diluents

                                18.10.7.2 Temperature Recording

                                18.10.7.3 Test Animals

                                18.10.7.4 Procedure

                                18.10.7.5 Test Interpretation and Continuation

18.11 Hemocompatibility

18.12 Emetic Responses

 

Chapter 19:         Pharmacokinetics and Toxicokinetics in Drug Safety Evaluation

 

19.1 Introduction

19.2 Regulations

19.3 Principles

                19.3.1 Preliminary Work

                19.3.2 Absorption

                                19.3.2.1 Absorption from the Pulmonary System

                                19.3.2.2 Absorption across the Skin

19.3.2.3 Parameters Controlling Absorption

                19.3.3 Distribution          

                                19.3.3.1 Protein Binding

                                19.3.3.2 Water Solubility

                                19.3.3.3 Volume of Distribution

                19.3.4 Metabolism/Biotransformation  

                                19.3.4.1 Metabolic Activation

                                19.3.4.2 Induction of P450 Metabolism and Isoenzymes

                                19.3.4.3 Species Differences

                19.3.5 Excretion

                                19.3.5.1 Urine

                                19.3.5.2 Feces

                                19.3.5.3 Expired Air

19.4 Pharmacokinetics

19.5 Laboratory Methods

                19.5.1 Analytical Methods

                                19.5.1.1 Instrumental Methods

                                19.5.1.2 Radiochemical Methods

                                19.5.1.3 Immunoassay Methods

19.6 Sampling Methods and Intervals

                19.6.1 Blood

                                19.6.2 Excreta

                                19.6.3 Bile          

                19.6.4 Expired Air

                19.6.5 Milk

                                19.6.5.1 Sampling Interval

19.7 Study Types

                19.7.1 Whole-Body Autoradiography

                19.7.2 Mass Balance Studies

                                19.7.2.1 In Vitro Studies

19.8 Analysis of Data

19.8.1 Use of Data from Metabolism and Pharmacokinetic Studies

                19.8.1.2 Design of Toxicity Studies

                19.9 Noncompartmental Analysis

                19.10 Physiologically Based Pharmacokinetic (PBPK) Modeling

19.11 Biologically Derived Materials

                19.11.1 Immunoassay Methods

                                19.11.1.1 Metabolism and Elimination

19.12 Points to Consider

 

Chapter 20:         Safety Pharmacology

20.1 Regulatory Requirements

20.2 Study Designs and Principles

                20.3 Organ System-Specific Tests

20.3.1 General Considerations in Selection and Design of Safety Pharmacology Studies

20.3.2 Studies on Metabolites, Isomers, and Finished Products

                20.4 Cardiovascular

20.4.1 Hemodynamics, ECG, and Respiration in Anesthetized Dogs or Primates

20.4.2 Cardiac Conduction Studies

20.4.3 Conscious Dog, Primate, or Minipig Telemetry Studies

20.4.4 Six-Lead ECG Measurement in the Conscious Dog and Minipig

20.4.5 Systems for Recording Cardiac Action Potentials

20.4.6 Special Case (and Concern): QT Prolongation

20.4.7 Some Specific Techniques which can be Employed

                20.4.7.1 Cloned Human Potassium Channels

                20.4.7.2 Cardiac Action Potential in Vitro: Purkinje Fibers

                20.4.7.3 Monophasic Action Potential in Anesthetized Guinea Pigs

                20.4.7.4 ECG by Telemetry in Conscious Dogs or Primates

                20.4.7.5 Hemodynamics and ECG in Anesthetized or Conscious Dogs or Primates

20.4.8 Relevance of hERG to QT Prolongation

                20.4.8.1 Expression and Recording Systems

                20.5 Central Nervous System

                                20.5.1 Isolated Tissue Assays

                                20.5.2 Electrophysiology Methods

                                20.5.3 CNS Function: Electroencephalography

                                20.5.4 Neurochemical and Biochemical Assays

                20.6 Respiratory/Pulmonary System

                                20.6.1 Design of Respiratory Function Safety Studies

                                20.6.2 Capnography

                20.7 Secondary Organ System

20.7.1 Gastric Emptying Rate and Gastric pH Changes:  A New Model

                20.8 Renal Function Tests

                20.9 Summary

 

Chapter 21:         Special Concerns for the Preclinical Evaluation of Biotechnology Products

21.1 Regulation

21.2 Preclinical Safety Assessment

21.3 Recombinant DNA Technology

                21.3.1 General Safety Issues

                21.3.2 Specific Toxicological Concerns

21.4 Immunogenicity/Allergenicity

21.5 Monoclonal Antibody Therapeutics

21.5.1 Toxicological Concerns with Monoclonal Antibodies

21.6 Bioprocess Technology

                21.7 Gene Therapy Products

                                21.7.1 Vectors

                21.7.2 Studies to Support the First Dose in Man

                21.7.3 Distribution of the Gene and Gene Product

                                21.7.4 Studies to Support Multiple Doses in Humans

21.7.5 Unnecessary Studies

21.7.6 Ex Vivo Procedures

21.7.7 Change of Gene or Vector

21.7.8 Change of Route

21.7.9 Insertional Mutagenesis

21.8 Vaccines

21.8.1 Approaches to Vaccination

21.8.2 Genetic Engineering and Vaccine Development

                21.8.2.1 DNA/Oligonucleotide Hybridization

                21.8.2.2 Hybrid Selection and Cell-Free Translation

                21.8.2.3 Expression Cloning

                21.8.2.4 Expression of Potential Vaccine Antigens

21.9 Special Challenges

21.9.1 Purity and Homology

21.9.2 Immunogenicity

21.10 Planning a Safety Evaluation Program

21.10.1 The Producing System

21.10.2 The Process

21.10.3 The Product

21.10.4 Biology of Bioengineered Products

21.10.5 Animal Models

21.10.6 Study Design

21.10.7 Frequency and Route of Administration

21.10.8 Duration

21.10.9 Special Toxicity Testing

21.10.10 Program Design Considerations

21.11 Challenges: Biosimilars

 

Chapter 22:         Safety Assessment of Inhalant Drugs and Dermal Route Drugs

22.1 Inhaled Therapeutics

22.2 The Pulmonary System       

22.3 Penetration and Absorption of Inhaled Gases and Vapors

22.4 Deposition of Inhaled Aerosols

22.5 Absorption and Clearance of Inhaled Aerosols

22.6 Pharmacokinetics and Pharmacodynamics of Inhaled Aerosols

22.7 Methods for Safety Assessment of Inhaled Therapeutics

22.8 Parameters of Toxicity Evaluation

22.8.1 The Inhaled “Dose”

22.8.2 The Dose–Response Relationship

22.8.3 Exposure Concentration versus Response

22.8.4 Product of Concentration and Duration (Ct) versus Responses

22.8.5 Units for Exposure Concentration

22.9 Inhalation Exposure Techniques

22.10 The Utility of Toxicity Data

22.11 Formulation and Potential Mucosal Damage

22.11.1 Methods to Assess Irritancy and Damage

22.12 Therapeutic Drug Delivery by the Dermal Route

 

Chapter 23:         Special Case Products: Imaging Agents

23.1 Introduction

23.2 Imaging Agents

23.2.1 Contrast Agents

23.2.2 Diagnostic Radiopharmaceuticals

23.2.3 Medical Imaging Agent Characteristics Relevant to Safety

                23.2.3.1 Mass Dose

23.2.3.2 Route of Administration

23.2.3.3 Frequency of Use

23.2.3.4 Biological, Physical, and Effective Half-Lives

23.2.4 Performance of Nonclinical Safety Assessments

                23.2.4.1 Nonclinical Safety Assessments for Nonbiological Drug Products

                23.2.4.2 Diagnostic Radiopharmaceuticals (Nonbiological Products)

Chapter 24:         Special Case Products: Drugs for Treatment of Cancer

24.1 Introduction

24.1.1 Animal Models

                24.1.2 Statistical Analysis of Study Results

24.2 How Oncology is Different

24.3 Dose Conversions: Perspective

                24.3.1 The Use of the mg/m2 Dose Unit

                24.3.2 Calculations of Drug Dosages for Treatment

                24.3.3 Conversion of mg/kg BW Doses to Units of mg/m2

24.4 Dose Setting in Oncology and non-infectious Imminently Fatal Diseases: STD10 and HNSTD

24.4.1 Determination of first in human dose levels based on pivotal toxicology study data

Chapter 25:         Pediatric Product Safety Assessment (2006 Guidance, Including Juvenile Toxicology)

25.1 Introduction

25.1.1 Scope of Nonclinical Safety Evaluation

25.1.2 Timing of Juvenile Animal Studies in Relation to Clinical Testing

                25.1.2.1 Long-Term Exposure in Pediatric Subjects

                25.1.2.2 Short-Term Exposure in Pediatric Subjects

                25.1.2.3 Insufficient Clinical Data to Support Initiation of Pediatric Studies

25.2 Issues to Consider Regarding Juvenile Animal Studies

25.2.1 Developmental Stage of Intended Population

25.2.2 Evaluating Data to Determine When Juvenile Animal Studies Should Be Used

25.2.3 Considering Developmental Windows When Determining Duration of Clinical Use

25.2.4 Timing of Exposure

25.2.5 Selection of Study Models

25.3 General Considerations in Designing Toxicity Studies in Juvenile Animals

25.4 Study Designs and Considerations

Chapter 26:         Use of Imaging, Imaging Agents, and Radiopharmaceuticals in Nonclinical Toxicology

26.1 Introduction

26.1.1 Multimodality Imaging Techniques

26.1.2 Dynamic Molecular Imaging Techniques

26.2 X‐ray

26.2.1 Angiography

26.3 Positron Emission Tomography (PET)

26.4 Single‐photon Emission Computed Tomography (SPECT)

26.5 Computed Tomography (CT)

26.6 Magnetic Resonance Imaging (MRI)

26.7 Optical Imaging

26.8 Ultrasound

26.8.1 Echocardiography

26.9 Nanoparticle Contrast Agent

26.10 Radiopharmaceuticals

26.11 Applications of Preclinical Imaging in Laboratory Animals

26.11.1 Molecular Imaging as an ADME Platform in Drug Screen

26.11.2 Preclinical Imaging in Oncology

26.11.3 Preclinical Imaging of CNS Disease

26.11.4 Preclinical Imaging of Autoimmune Disease

26.11.5 Imaging Animal Model of Infectious Disease

26.11.6 Preclinical Imaging of Cardiac Disease

26.12 Nonclinical Safety Assessment for Imaging Agents

26.13 Radiopharmaceuticals

26.14 Nonclinical Late Radiation Toxicity Studies

26.14.1 Study Goals

26.15 Study Design

26.15.1 Good Laboratory Practices

26.15.2 Species Selection

26.15.3 Timing of Study

26.15.4 General Study Design

26.15.5 Dose Levels

26.15.6 Clinical Pathology

26.15.7 Necropsy and Histopathology

Chapter 27:         Occupational Toxicology in the Pharmaceutical Industry                                               

27.1 Introduction

27.2 Occupational Toxicology versus Drug Safety Evaluation

27.3 Regulatory Pressures in the United States and the European Community

27.4 Organizational Structure

27.5 Activities

27.5.1 Data Evaluation and Dissemination

27.5.2 Data Development

27.5.3 Occupational Exposure Limits (OELs)

27.5.4 Hazard Assessment

27.5.5 Employee Training

27.6 Conclusion

 

Chapter 28:         Strategy and Phasing for Nonclinical Drug Safety Evaluation in the Discovery and Development of Pharmaceuticals

28.1 Introduction

28.2 Regulatory Requirements

28.3 Essential Elements of Project Management

28.4 Screens: Their Use and Interpretation in Safety Assessment

28.4.1 Characteristics of Screens

28.5 Strategy and Phasing

28.6 Critical Considerations

28.7 Special Cases in Safety Assessment

28.8 Potential Market Withdrawal Issues

28.9 Summary

Chapter 29:         The Application of In Vitro Techniques in Drug Safety Assessment

29.1 Introduction

29.2 In Vitro Testing in Pharmaceutical Safety Assessment

29.3 Defining Testing Objective

29.3.1 Objectives behind Data Generation and Utilization

29.4 Test Systems: Characteristics, Development, and Selection

29.5 In Vitro Models

29.6 Local Tissue Tolerance

29.6.1 Ocular Irritation

29.6.2 Dermal Irritation

29.6.3 Irritation of Parenterally Administered Pharmaceuticals

29.6.4 Sensitization and Photosensitization

29.6.5 Phototoxicity and Photosensitization

29.6.6 Pyrogenicity

29.6.7 Developmental Toxicity

29.6.8 Target Organ Toxicity Models

29.7 In Silico Methods

29.8 The Final Frontier and Barrier: Regulatory Acceptance

29.9 Summary

 

Chapter 30:         Evaluation of Human Tolerance and Safety in Clinical Trials: Phase I and Beyond

30.1 The Pharmaceutical Clinical Development Process and Safety

30.1.1 Pharmacokinetics

30.1.1.1 Relating the Time Course of Plasma Concentrations to the Time Course of Effect

30.1.2 Safety of Clinical Trial Subjects

                30.1.2.1 Regulations

                30.1.2.2 Increased Frequency Reports

                30.1.2.3 Reporting Forms

                30.1.2.4 Definitions

30.1.2.5 Time Frames

30.1.2.6 Continuous Safety Monitoring

30.1.2.7 Sponsor Pharmacovigilance

30.2 Limitations on/of Clinical Trials

30.3 The Clinical Trial Process

30.3.1 Development of an Application Unrelated to Original Approved Use

                30.3.1.1 Special Considerations

30.4 Institutional Review Boards (IRBS)/Ethics Committees in the Clinical Trial Process

30.4.1 Legal Authority and Responsibilities for IRBs and ECs

30.4.2 Duties of IRBs

30.4.3 Informed Consent

30.5 Drug Formulations and Excipients

30.5.1 Route of Administration

30.6 Phase I Designs

30.6.1 First Administration: Single Dose Escalating (SDE)

30.6.2 First Administration in Humans: Multiple Dose Escalating (MDE)

30.6.2.1 Number of Subjects

30.7 Clinical Trial Safety Indicators

30.7.1 Overall Approach to Assessing Safety

                30.7.1.1 Choosing Safety Parameters

                30.7.1.2 Measuring Safety Parameters

30.7.1.3 Parameters That Measure either Safety or Efficacy

30.7.1.4 Appropriateness of Each Parameter for the Clinical Trial and Patient

                30.7.2 Precautions         

                                30.7.2.1 Summary of Tests

                                30.7.2.2 Choosing Laboratory Tests

                                30.7.2.3 Tests in Phase I

                                30.7.2.4 Tests in Later Phases

                                30.7.2.5 Tests in Medical Practice

                                30.7.2.6 Less Commonly Used Methods

                30.7.2.7 Identifying the Most Important Laboratory Analytes to Monitor in a Clinical Trial

30.7.2.8 Uses of Specific Laboratory Tests to Discover, Confirm, and/or Exclude a Disease

30.7.2.9 Hematology

30.7.3 Clinical Chemistry

                30.7.3.1 Drug Levels in Plasma

                30.7.3.2 Total Blood That May be Taken from Patients

30.7.4 Urinalysis

30.7.5 Urine Screens

                30.7.5.1 Type of Container to Be Used

                30.7.5.2 Use of International System Units

30.7.6 Identifying New Diagnostic Laboratory Tests

30.7.7 Ophthalmological Examination

30.7.8 Dermatological Examinations

30.7.9 Cardiovascular Safety

30.7.10 Deaths in Clinical Trials

30.7.11 Behavioral Rating Scales, Performance, Personality, and Disability Tests

30.7.12 Adult Behavioral Rating Scales

                30.7.12.1 Anxiety Status Inventory

30.7.12.2 Back Depression Inventory

30.7.12.3 Brief Psychiatric Rating Scale

30.7.12.4 Carroll Rating Scale for Depression

30.7.12.5 Clinical Global Impressions

30.7.12.6 Clyde Mood Scale

30.7.12.7 Covi Anxiety Scale

30.7.12.8 Crichton Geriatric Rating Scale

30.7.12.9 Depression Status Inventory

30.7.12.10 Hamilton Anxiety Scale

30.7.12.11 Hamilton Depression Scale

30.7.12.12 Hopkins Symptom Checklist

30.7.12.13 Inpatient Multidimensional Psychiatric Scale

30.7.12.14 Nurses’ Observation Scale for Inpatient Evaluation

30.7.12.15 Plutchik Geriatric Rating Scale

30.7.12.16 Profile of Mood States

30.7.12.17 Sandoz Clinical Assessment-Geriatric

30.7.12.18 Self-Report Symptom Inventory

30.7.12.19 Wittenborn Psychiatric Rating Scale

30.7.12.20 Zung Self-Rating Anxiety Scale

30.7.12.21 Zung Self-Rating Depression Scale

30.7.13 Pediatric Behavioral Rating and Diagnostic Scales

30.7.13.1 Children’s Behavior Inventory

30.7.13.2 Children’s Diagnostic Classification

30.7.13.3 Children’s Diagnostic Scale

30.7.13.4 Children’s Psychiatric Rating Scale

30.7.13.5 Clinical Global Impression

30.7.13.6 Conners Parent Questionnaire

30.7.13.7 Conners Parent/Teacher Questionnaire

30.7.13.8 Conners Teacher Questionnaire

30.7.13.9 Devereux Child Behavior Rating Scale

30.7.13.10 Devereux Elementary School Behavior Rating Scale

30.7.14 Psychometric and Performance Tests

30.7.14.1 Bender-Gestalt Test

30.7.14.2 Conceptual Clustering Memory Test

30.7.14.3 Digit Symbol Substitution Test

30.7.14.4 Embedded Figures Test

30.7.14.5 Frostig Developmental Test of Visual Perception

30.7.14.6 Goodenough–Harris Figure-Drawing Test

30.7.14.7 Peabody Picture Vocabulary Test

30.7.14.8 Porteus Mazes

30.7.14.9 Reaction Time

30.7.14.10 Vigilance Tests

30.7.14.11 Wechsler Adult Intelligence Scale

30.7.14.12 Wechsler Intelligence Scale for Children

30.7.14.13 Wechsler Memory Scale

30.7.14.14 Wide Range Achievement Test

30.7.15 Personality Tests

30.8 Assessment of Unwanted Drug Effects

30.8.1 Separation of Adverse Reactions from Placebo Reactions

30.8.1.1 Base-Case Causality of Single-Event Adverse Drug Reactions

30.9 Recent Changes in Safety Related Requirements for Initial Clinical Trials

 

Chapter 31:         Postmarketing Safety Evaluation: Monitoring, Assessing, and Reporting of Adverse Drug Responses (ADRs)

31.1 Causes of Safety Withdrawals

31.2 Regulatory Requirements

31.2.1 The 15‐Day Report versus the US Periodic Report

31.3 Management of ADR and ADE Data

31.3.1 Sources of Data

31.3.2 Clinical Trials,

31.3.3 Post-marketing Surveillance Studies

31.3.4 Spontaneous Reports

31.3.5 Literature

31.3.6 Searching for ADRs in the Literature

31.3.7 Information Required for Reports

31.3.8 Adverse Drug Reaction Forms and Form Design

31.3.9 Computerization of Drug Safety Data: Data Collection and Input

31.3.10 Medical and Drug Terminology

31.3.11 Dictionaries

31.3.12 Medical Term Coding Dictionaries

31.3.13 Medical Dictionary for Regulatory Activities

                                                31.3.13.1 FDA

31.3.13.2 European Union

31.3.13.3 Japan

31.3.14 Periodic Reports

31.4 Causality Assessment

31.4.1 Aims of Causality Assessment

31.5 Courses of Corrective Action

31.6 Legal Consequences of Safety Withdrawal

31.6.1 FDA Tools for Risk Management

                31.6.1.1 The Regulatory Pyramid

31.6.2 Tier 1: Mandatory Studies

31.6.3 Tier 2: Labeling and Assessment

31.6.4 Tier 3: Enhanced Communication

31.6.5 Tier 4: Safe Use Restriction Defined by Provider

31.6.6 Tier 5: Safe Use Restriction Defined by Patient

Chapter 32:         Statistics in Pharmaceutical Safety Assessment

32.1 Introduction

32.1.1 Bias and Chance

32.1.2 Hypothesis Testing and Probability (p) Values

32.1.3 Multiple Comparisons

32.1.4 Estimating the Size of the Effect

32.1.5 Functions of Statistics

32.1.6 Descriptive Statistics

32.2 Experimental Design

32.2.1 Choice of Species and Strain

32.2.2 Sampling

32.2.3 Dose Levels

32.2.4 Number of Animals

32.2.5 Duration of the Study

32.2.6 Stratification

32.2.7 Randomization

32.2.8 Adequacy of Control Group

32.3 Data Recording

32.4 Generalized Methodology Selection

32.5 Statistical Analysis: General Considerations

32.5.1 Variables to Be Analyzed

32.5.2 Combination of Observations (Such as Pathological Conditions)

32.5.3 Taking Severity into Account

32.5.4 Using Simple Methods Which Avoid Complex Assumptions

32.5.5 Using All the Data

32.5.6 Combining, Pooling, and Stratification

32.5.7 Trend Analysis, Low‐Dose Extrapolation, and NOEL Estimation

32.5.8 Need for Age Adjustment

32.5.9 Need to Take Context of Observation into Account

32.5.10 Experimental and Observational Units

32.5.11 Missing Data

32.5.12 Use of Historical Control Data

32.5.13 Methods for Data Examination and Preparation

32.5.14 Scattergram

32.5.15 Bartlett’s Test for Homogeneity of Variance

32.5.16 Statistical Goodness‐of‐Fit Tests

32.5.17 Randomization

32.5.18 Transformations

32.5.19 Exploratory Data Analysis

32.6 Hypothesis Testing of Categorical and Ranked Data

32.6.1 Fisher’s Exact Test

32.6.2 2 × 2 Chi‐Square

32.6.3 R × C Chi‐Square

32.6.4 Wilcoxon Rank‐Sum Test

32.6.5 Distribution‐Free Multiple Comparison

32.6.6 Mann–Whitney U Test

32.6.7 Kruskal–Wallis Nonparametric ANOVA

32.6.8 Log‐Rank Test

32.7 Hypothesis Testing: Univariate Parametric Tests

32.7.1 Student’s t‐Test (Unpaired t‐Test)

32.7.2 Cochran t‐Test

32.7.3 F‐Test

32.7.4 Analysis of Variance (ANOVA)

32.7.5 Post Hoc Tests

32.7.6 Duncan’s Multiple Range Test

32.7.7 Groups with Equal Number of Data (N1 = N2)

32.7.8 Groups with Unequal Number of Data (N1 ≠ N2)

32.7.9 Scheffe’s Multiple Comparisons

32.7.10 Dunnett’s t‐Test

32.7.11 Williams’ t‐Test

32.7.12 Analysis of Covariance

32.7.13 Modeling

32.7.14 Linear Regression

32.7.15 Probit/Log Transforms and Regression

32.7.16 Nonlinear Regression

32.7.17 Correlation Coefficient

32.7.18 Kendall’s Coefficient of Rank Correlation

32.7.19 Trend Analysis

32.8 Methods for the Reduction of Dimensionality

32.8.1 Classification

32.8.2 Statistical Graphics

32.8.3 Multidimensional and Nonmetric Scaling

32.8.4 Cluster Analysis

32.8.5 Fourier or Time Analysis

32.8.6 Life Tables

32.9 Meta‐Analysis

32.9.1 Selection of the Studies to Be Analyzed

32.9.2 Pooled (Quantitative) Analysis

32.9.3 Methodological (Qualitative) Analysis

32.10 Bayesian Inference

32.10.1 Bayes’ Theorem and Evaluation of Safety Assessment Studies

32.10.2 Bayes’ Theorem and Individual Animal Evaluation

32.11 Data Analysis Applications in Safety Assessment Studies

32.11.1 Body and Organ Weights

32.11.2 Clinical Chemistry

32.11.3 Hematology

32.11.4 Histopathological Lesion Incidence,

32.11.5 Carcinogenesis

Chapter 33:         Combination Products: Drugs and Devices

33.1 Combination Products

33.1.1 Historical Background

33.1.2 Future Trends

33.1.2.1 Device Programs That CDER and CDRH Each will Administer

33.1.2.2 Coordination

33.1.2.3 Submissions

33.1.2.4 Center Jurisdiction

33.1.2.5 General Criteria Affecting Drug/Device Determination

 

Chapter 34:         Qualification of Impurities, Degradants, Residual Solvents, Metals, and Leachables in Pharmaceuticals

32.1 Impurities

32.2 Residual Solvents

32.3 Extractables and Leachables

32.4 Residual Metals and Elements

 

Chapter 35:         Tissue, Cell, and Gene Therapy

35.1 Safety Assessment of Cell Therapy (CT) Products

35.1.1 Recommendations for General Preclinical Program Design

35.1.2 Model Species Selection

35.1.3 Selection of Animal Models of Disease/Injury

35.1.4 Information Describing Limitations of Potential Animal Model(s)

35.1.5 Information Supporting the Choice of Animal Model(s)

35.1.6 Proof‐of‐Concept (POC) Studies

35.1.7 Toxicology Studies

                35.1.7.1 Primary Considerations for Toxicology Study Design

                35.1.7.2 Secondary Considerations for Toxicology Study Design

35.1.8 Product Delivery Considerations

35.1.9 Study Designs

35.1.10 CT Products with Implantable Scaffolds

35.2 Nonclinical Safety Assessment of Gene Therapy Products (GTPS)

35.2.1 CBER

35.2.2 NIH

35.2.3 Study Designs

35.2.4 Ex Vivo Genetically Modified Cells

35.2.5 Biodistribution Considerations

35.3 Definitions

 

Chapter 36:  Adverse Outcome Pathways in Drug Safety Assessment and Drug Development

                36.1 Introduction

                36.2 Initial Steps

                36.3 Test Article – Confirming Identity and Stability

                36.4 Formulation

                36.5 Test Species – Animal Models

35.6 Dose Level Selection and Dosing Errors

36.7 Poor Planning

36.8 Pay Attention to the Regulatory Clock and Changes in Requirements and Guidelines

36.9 Most Advances in Safety Assessment Are Small

 

Appendices

A: Selected Regulatory and Toxicological Acronyms

B: Definition of Terms and Lexicon of Observations in Nonclinical (Animal) Studies

C: Notable Regulatory Internet Addresses

D: Glossary of Terms Used in the Clinical Evaluation of Therapeutic Agents

E: Common Vehicles for the Non-Clinical Evaluation of Therapeutic Agents

F: Global Directory of Contract Toxicology Labs

 

 

INDEX 879

Supplemental Materials

What is included with this book?

The New copy of this book will include any supplemental materials advertised. Please check the title of the book to determine if it should include any access cards, study guides, lab manuals, CDs, etc.

The Used, Rental and eBook copies of this book are not guaranteed to include any supplemental materials. Typically, only the book itself is included. This is true even if the title states it includes any access cards, study guides, lab manuals, CDs, etc.

Rewards Program