Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulations

Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulations
Автор книги: id книги: 2170470     Оценка: 0.0     Голосов: 0     Отзывы, комментарии: 0 22620,7 руб.     (246,47$) Читать книгу Купить и скачать книгу Электронная книга Жанр: Медицина Правообладатель и/или издательство: John Wiley & Sons Limited Дата добавления в каталог КнигаЛит: ISBN: 9781119497790 Скачать фрагмент в формате   fb2   fb2.zip Возрастное ограничение: 0+ Оглавление Отрывок из книги

Реклама. ООО «ЛитРес», ИНН: 7719571260.

Описание книги

Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulations The first book dedicated to the emerging field of physiologically based pharmacokinetic modeling (PBPK) Now in its second edition, Physiologically Based Pharmacokinetic (PBPK) Modelling and Simulations: Principles, Methods, and Applications in the Pharma Industry remains the premier reference book throughout the rapidly growing PBPK user community. Using clear and concise language, author Sheila Annie Peters connects theory with practice as she explores the vast potential of PBPK modeling for improving drug discovery and development. This fully updated new edition covers key developments in the field of PBPK modelling and simulations that have emerged in recent years. A brand-new section provides case studies in different application areas of PBPK modelling, including drug-drug interaction, genetic polymorphism, renal impairment, and pediatric extrapolation. Additional chapters address topics such as model-informed drug development (MIDD) and expose readers to a wide range of current applications in the field. Throughout the book, substantially revised chapters simplify complex topics and offer a balanced view of both the opportunities and challenges of PBPK modelling. Providing timely and comprehensive coverage of one of the most exciting new areas of pharmaceutical science, this book: Describes the principles behind physiological modeling of pharmacokinetic processes, inter-individual variability, and drug interactions for small molecule drugs and biologics Features a wealth of new figures and case studies of the applications of PBPK modelling along the value chain in drug discovery and development Reflects the latest regulatory guidelines on the reporting of PBPK modelling analysis Includes access to a new companion website containing code, datasets, explanations of case examples in the text, and discussion of key developments in the field Contains a brief overview of the field, end-of-chapter keywords for easy reference, and an extensive bibliography Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulations: Principles, Methods, and Applications in the Pharmaceutical Industry, Second Edition is an indispensable ­single-volume resource for beginning and intermediate practitioners across the pharmaceutical sciences in both industry and academia.

Оглавление

Sheila Annie Peters. Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulations

Table of Contents

List of Tables

List of Illustrations

Guide

Pages

PHYSIOLOGICALLY BASED PHARMACOKINETIC (PBPK) MODELING AND SIMULATIONS. Principles, Methods, and Applications in the Pharmaceutical Industry

PREFACE TO THE SECOND EDITION

PREFACE TO THE FIRST EDITION

ACKNOWLEDGMENTS

ABOUT THE COMPANION WEBSITE

1 A REVIEW OF PHARMACOKINETIC AND PHARMACODYNAMIC PRINCIPLES. CONTENTS

1.1 INTRODUCTION

1.2 PHARMACOKINETIC PRINCIPLES. 1.2.1 Routes of Drug Administration

1.2.2 Intravenous Bolus

1.2.2.1 Zero‐ and First‐Order Kinetics

1.2.2.2 Clearance, Volume of Distribution, Half‐life, and AUC

1.2.3 Plasma Protein Binding and Blood–Plasma Ratio

1.2.4 Hepatic, Renal, and Biliary Clearances

1.2.4.1 Hepatic Clearance

1.2.4.2 Hepatic Extraction

1.2.4.3 Renal Clearance

1.2.4.4 Biliary Clearance

1.2.5 Extravascular (Subcutaneous, Intramuscular, and Per Oral) Absorption

1.2.6 Absorption from Solid Dosage Forms

Box 1.1

1.2.7 Role of Transporters in ADME

1.2.8 Linear and Non‐Linear Pharmacokinetics

1.2.9 Intravenous Infusion, Repeated Dosing, Steady State Kinetics, and Accumulation

1.2.10 Active Metabolite and Prodrug Kinetics. 1.2.10.1 Active Metabolites

1.2.10.2 Prodrugs

1.2.10.3 Metabolite Kinetics

1.2.10.4 Limiting Conditions

1.2.10.5 Drug‐Metabolite Plasma Concentration Relationships After Single Drug Administration

1.2.10.6 Steady‐State Metabolite Kinetics

1.2.10.7 First‐Pass Metabolite Production

1.3 PHARMACOKINETIC VARIABILITY

1.4 PHARMACOKINETICS OPTIMIZATION IN DRUG DISCOVERY

1.5 PHARMACODYNAMIC PRINCIPLES

1.5.1 Pharmacological Targets and Drug Action

1.5.2 Functional Adaptation Processes

1.5.2.1 Desensitization, Tolerance, and Tachyphylaxis

1.5.3 Biomarkers, Surrogate Endpoints, and Clinical Endpoints

KEYWORDS

REFERENCES

2 A REVIEW OF DRUG–DRUG INTERACTIONS. CONTENTS

2.1 INTRODUCTION

2.2 DRUG INTERACTIONS MEDIATED BY ENZYMES AND TRANSPORTERS AT VARIOUS SITES

2.3 FACTORS AFFECTING DDI

2.4 IN VITRO METHODS TO EVALUATE DRUG–DRUG INTERACTIONS

2.4.1 Candidate Drug as a Potential Perpetrator

2.4.2 Candidate Drug as a Potential Victim of Inhibition

2.5 SOURCES OF UNCERTAINTY

2.6 THERAPEUTIC PROTEIN–DRUG INTERACTION

KEYWORDS

REFERENCES

3 MODELING PHARMACOKINETICS, PHARMACODYNAMICS, AND DRUG INTERACTIONS. CONTENTS

3.1 INTRODUCTION

3.2 MODELING PHARMACOKINETICS

3.2.1 Compartmental Modeling of Linear and Nonlinear Pharmacokinetics (Enzyme and/or Transporter Capacity Limitation as Well as Target‐Mediated Drug Disposition) 3.2.1.1 Linear PK

3.2.1.2 Nonlinear PK (Enzyme and/or Transporter Capacity Limitation)

3.2.1.3 Nonlinear PK (Target‐Mediated Drug Disposition, TMDD) (Gibiansky et al. 2008)

3.2.2 Population Pharmacokinetics

3.3 PHARMACOKINETICS/PHARMACODYNAMICS AND PK/EFFICACY (EXPOSURE/RESPONSE) MODELING

3.3.1 PK/PD Models for Direct Effect: Sigmoid Emax Model

3.3.2 PK/PD Models for Direct Effect: Classical Receptor Theory

3.3.3 PK/PD Models Accommodating Delayed Pharmacological Response

3.3.4 PK/PD Models Accommodating Functional Adaptation Leading to Nonlinearity in Pharmacological Response with Respect to Time

3.3.5 PK/Efficacy Modeling

3.3.6 Translation of PK/PD and PK/Efficacy Modeling to Human

3.3.7 Average, Minimum, and Maximum Steady‐State Concentrations

Box 3.1 Decision Tree for Choice of PK Metric (Maximum/Minimum/Average Steady State Concentration) and Extent and Duration of PD Modulation for Estimation of Human Dose

3.3.8 Estimation of Biologically Effective Dose in Human

3.3.9 Therapeutic Window

3.3.10 Static Models for Drug Interactions

3.3.10.1 Reversible Inhibition

3.3.10.2 Time‐Dependent Inhibition

3.3.10.3 Induction

3.4 PHYSIOLOGICALLY BASED PHARMACOKINETIC (PBPK) MODELING AND ITS INTEGRATION WITH PHARMACODYNAMICS AND EFFICACY MODELS

3.4.1 PK Modeling Compartmental vs PBPK

3.4.2 PK Variability: Population PK (popPK) Modeling vs PBPK

3.4.3 Integration of PBPK with PD, Quantitative Systems Pharmacology (QSP) Models or Quantitative Systems Toxicology and Safety (QSTS)

3.4.4 PBPK Models to Evaluate Drug–Drug Interactions

3.4.4.1 Intrinsic Clearance of Victim (V) in the Absence of Inhibitor or Inducer

3.4.4.2 Intrinsic Clearance of Victim (V) in the Presence of Inhibitor

3.4.4.3 Time‐Dependent Changes in the Abundance of an Enzyme Isoform Inhibited by a MBI

3.4.5 DDI Risk Assessment with PBPK vs Static Models

Box 3.2 Evaluation of NME as Victim and Perpetrator of CYP Inhibition and Induction (USFDA 2020a). Adapted from USFDA, 2020b

Box 3.3 Evaluation of Metabolite as Victim of CYP Inhibition and Perpetrator of CYP and Transporter Inhibition (USFDA 2020a). Adapted from USFDA 2020b

Box 3.4 Evaluation of NME as Victim and Perpetrator of Transporter Inhibition (USFDA 2020a). Adapted from USFDA 2020b

NME as a Victim of Transporter Inhibition

NME as an Inhibitor of Transporters

KEYWORDS

REFERENCES

4 PHYSIOLOGICAL MODEL FOR ABSORPTION. CONTENTS

4.1 INTRODUCTION

4.2 DRUG ABSORPTION AND GUT BIOAVAILABILITY

4.2.1 Solubility and Dissolution Rate

4.2.2 Permeability: Transcellular, Paracellular, and Carrier‐Mediated Pathways

4.2.3 Barriers to Membrane Transport – Luminal Degradation, Efflux, and Gut Metabolism

4.3 FACTORS AFFECTING DRUG ABSORPTION AND GUT BIOAVAILABILITY

4.3.1 Physiological Factors Affecting Oral Drug Absorption and Species Differences in Physiology

4.3.2 Compound‐Dependent Factors

4.3.3 Formulation‐Dependent Factors

4.4 IN SILICO PREDICTIONS OF PASSIVE PERMEABILITY AND SOLUBILITY. 4.4.1 In Silico Models for Permeability

4.4.2 In Silico Models for Solubility

4.5 MEASUREMENT OF PERMEABILITY, SOLUBILITY, LUMINAL STABILITY, EFFLUX, INTESTINAL METABOLISM

4.5.1 In Vitro, In Situ, and In Vivo Models for Effective Permeability

4.5.2 Measurement of Thermodynamic or Equilibrium Solubility

4.5.3 Luminal Stability

4.5.4 Efflux

4.5.5 In Vitro Models for Gut Metabolism and Estimation of Fraction Escaping Gut Metabolism

4.6 ABSORPTION MODELING

KEYWORDS

REFERENCES

5 PHYSIOLOGICAL MODEL FOR DISTRIBUTION. CONTENTS

5.1 INTRODUCTION

5.2 FACTORS AFFECTING TISSUE DISTRIBUTION OF XENOBIOTICS

5.2.1 Physiological Factors and Species Differences in Physiology

5.2.1.1 Blood Flow Rate

5.2.1.2 Membrane Permeability

5.2.1.3 Transporters

5.2.1.4 Tissue Compositions

5.2.1.5 Binding to Plasma Proteins and Tissue Components

5.2.2 Compound‐Dependent Factors

5.3 IN SILICO MODELS OF TISSUE PARTITION COEFFICIENTS

5.4 MEASUREMENT OF PARAMETERS REPRESENTING THE RATE AND EXTENT OF TISSUE DISTRIBUTION

5.4.1 Assessment of Rate and Extent of Brain Penetration

5.5 PHYSIOLOGICAL MODEL FOR DRUG DISTRIBUTION

5.6 DRUG CONCENTRATIONS AT THE SITE OF ACTION

KEYWORDS

REFERENCES

6 PHYSIOLOGICAL MODELS FOR DRUG METABOLISM AND EXCRETION. CONTENTS

6.1 INTRODUCTION

6.2 FACTORS AFFECTING DRUG METABOLISM AND EXCRETION OF XENOBIOTICS

6.3 MODELS FOR HEPATOBILIARY AND RENAL EXCRETION. 6.3.1 In Silico Models

6.3.2 In Vitro Models for Hepatic Metabolism

6.3.3 In Vitro Models for Transporters

6.3.3.1 Efflux Transporters

6.3.3.2 Uptake Transporters

6.4 PHYSIOLOGICAL MODELS

6.4.1 Hepato‐Biliary Elimination of Parent Drug and Metabolites

6.4.1.1 Parent Drug. 6.4.1.1.1 INTERSTITIAL (I) LIVER (LI) COMPARTMENT

6.4.1.1.2 INTRACELLULAR (IC) LIVER COMPARTMENT

6.4.1.2 Metabolite. 6.4.1.2.1 INTRACELLULAR LIVER COMPARTMENT

6.4.1.2.2 SYSTEMIC (SYS) CIRCULATION

6.4.2 Renal Excretion

6.4.2.1 Kidney. 6.4.2.1.1 VASCULAR (V)

6.4.2.1.2 INTERSTITIAL (I)

6.4.2.1.3 CHANGES IN VOLUME OF WATER ALONG THE TUBULE LUMEN

6.4.2.1.4 TUBULE LUMEN

6.4.2.1.5 INTRACELLULAR

REFERENCES

7 GENERIC WHOLE‐BODY PHYSIOLOGICALLY BASED PHARMACOKINETIC MODELING. CONTENTS

7.1 INTRODUCTION

7.2 STRUCTURE OF A GENERIC PHYSIOLOGICALLY‐BASED PHARMACOKINETIC (PBPK) MODEL

7.3 SOMATIC COMPARTMENTS. 7.3.1 Lungs (LU)

7.3.2 Arterial Blood (ART)

7.3.3 Venous Blood (VEN)

7.3.4 Stomach (ST)

7.3.5 Gut (GU)

7.4 MODEL ASSUMPTIONS

7.5 PBPK SOFTWARE

REFERENCES

8 PBPK MODELING OF BIOTHERAPEUTICS. CONTENTS

8.1 INTRODUCTION

8.2 THERAPEUTIC PROTEINS. 8.2.1 Peptides and Proteins

8.2.2 Antibodies and Antibody‐Based Therapies

8.3 PHARMACOKINETICS OF THERAPEUTIC PROTEINS

8.3.1 Absorption

8.3.2 Renal Elimination

8.3.3 Immunogenicity

8.3.4 PEGylation

8.3.5 Transport by Convective and Transcytotic Extravasation

8.3.6 Catabolic Elimination (Proteolysis)

8.3.7 FcRn‐Mediated Protection of IgGs Against Catabolism in FcRn‐Rich Cells

8.3.8 Distribution and lymphatic elimination

8.3.9 Target‐Mediated Drug Disposition and Receptor‐Mediated Endocytosis

8.4 PBPK MODELING OF MONOCLONAL ANTIBODIES. 8.4.1 Full PBPK Model for Monoclonal Antibodies

8.4.1.1 Plasma (PL)

8.4.1.2 Lung (LU) 8.4.1.2.1 VASCULAR SPACE

8.4.1.2.2 ENDOSOMAL SPACE

8.4.1.3 Interstitial Space

8.4.1.4 Other Organs (OT) and Tumor (TU) 8.4.1.4.1 VASCULAR SPACE

8.4.1.4.2 ENDOSOMAL SPACE

8.4.1.4.3 INTERSTITIAL SPACE

8.4.2 Minimal PBPK Model for Monoclonal Antibodies

8.5 APPLICATIONS OF PBPK MODELING OF MONOCLONAL ANTIBODIES. 8.5.1 Pharmacokinetic Scaling

8.5.2 PBPK Integration with Pharmacodynamics of Monoclonal Antibodies

KEYWORDS

REFERENCES

9 UNCERTAINTY AND POPULATION VARIABILITY. CONTENTS

9.1 INTRODUCTION

9.2 DISTINGUISHING UNCERTAINTY AND VARIABILITY

9.3 SOURCES OF UNCERTAINTY IN DRUG‐RELATED PARAMETERS

9.4 SOURCES OF VARIABILITY IN SYSTEM PARAMETERS

9.5 HANDLING POPULATION VARIABILITY. 9.5.1 A POSTERIORI and A PRIORI Approaches to Handling Population Variability

9.5.2 Correlations Between Parameters

9.6 UNCERTAINTY AND SENSITIVITY ANALYSIS

9.6.1 Local Sensitivity Analysis (One‐at‐a‐time (OAT) and Derivative‐based Methods)

9.6.2 Parameter Interactions and Global Sensitivity Analysis (GSA)

9.6.3 Global Sensitivity Analysis for Correlated Parameters (cGSA)

9.6.4 Applications of Sensitivity Analysis for PBPK Models

9.6.5 Limitations of Global Sensitivity Analysis

9.7 UNCERTAINTY AND POPULATION VARIABILITY IN CLINICAL EFFICACY AND SAFETY

KEYWORDS

REFERENCES

10 NONCLINICAL, CLINICAL, AND MODEL‐INFORMED DRUG DEVELOPMENT. CONTENTS

10.1 INTRODUCTION: AN OVERVIEW OF DIFFERENT PHASES OF DRUG DEVELOPMENT

10.2 NONCLINICAL DEVELOPMENT

10.2.1 Preclinical Pharmacology, PK/PD Modeling, and Human Dose Prediction

10.2.2 Safety and Toxicology Studies. 10.2.2.1 Safety Pharmacology Studies

10.2.2.2 Genotoxicity

10.2.2.3 Toxicology Studies

10.2.3 Studies with Radiolabeled Compound. 10.2.3.1 Metabolite Identification and Quantification (Isin et al., 2012)

10.2.3.2 Quantitative Whole‐Body Autoradiography (QWBA) (Solon, 2015)

10.2.3.3 Metabolite profiling

10.2.3.4 Preclinical ADME/Mass Balance (ADME/MB) Studies

10.2.3.5 Safety Testing of Drug Metabolites

10.2.3.6 Drug Interaction Potential of Metabolites of Significant Exposure or Pharmacological or Toxicological Activity

10.3 CLINICAL PHARMACOLOGY STUDIES

10.3.1 First‐in‐Human, Single, and Multiple Ascending Dose Studies

10.3.2 Biopharmaceutics – Absolute Oral Bioavailability and Bioequivalence Study

10.3.3 Food Effect Study

10.3.4 Organ (Hepatic and Renal) Impairment Study

10.3.5 Pediatric Assessment

10.3.6 Mass Balance Study

10.3.7 Drug Interaction Study

10.3.8 Pharmacogenomics Study

10.3.9 Thorough QT (TQT) and Concentration QT (C‐QT) Study

10.3.10 Immunogenicity Assays and Comparability Study for Biologics

10.3.11 Drug Labelling

10.4 CLINICAL DEVELOPMENT IN ONCOLOGY

10.5 FAST TRACK ROUTES TO ADDRESS UNMET MEDICAL NEED IN THE TREATMENT OF SERIOUS CONDITIONS

10.6 MODEL‐INFORMED DRUG DEVELOPMENT

10.7 PHYSIOLOGICALLY BASED PHARMACOKINETIC MODELS COMPLEMENTING CLINICAL PHARMACOLOGY STUDIES

10.8 PBPK IN ONCOLOGY

REGULATORY GUIDELINES

REFERENCES

11 OVERVIEW OF PBPK APPLICATIONS. CONTENTS

11.1 INTRODUCTION

11.2 PBPK APPLICATIONS FOR INTERNAL DECISIONS

11.3 PBPK APPLICATIONS FOR REGULATORY FILING

11.4 PBPK MODELING AND SIMULATIONS ALONG THE VALUE CHAIN

REFERENCES

12 APPLICATIONS OF HYPOTHESIS GENERATION AND TESTING WITH PBPK MODELS. CONTENTS

12.1 INTRODUCTION

12.2 HYPOTHESIS GENERATION AND TESTING WITH PBPK MODELS

12.2.1 Parameter Estimation from Intravenous Pharmacokinetic Profiles

12.2.2 Simulation of Oral PK Profile

12.2.3 Sensitivity Analysis

12.2.4 Verification of Hypotheses

12.2.5 Auto‐inhibition of Drug‐Metabolizing Enzymes, Uptake and Efflux Transporters

12.3 HYPOTHESIS GENERATION AND TESTING ALONG THE VALUE CHAIN

12.4 CONCLUSIONS

REFERENCES

13 APPLICATIONS OF PHYSIOLOGICALLY BASED PHARMACOKINETIC MODELS INTEGRATED WITH DRUG EFFECT MODELS (PBPK/PD) CONTENTS

13.1 INTRODUCTION: INTEGRATION OF PBPK WITH DRUG EFFECT MODELS

13.2 DOSING IN SPECIFIC POPULATIONS

13.3 PBPK/PD FOR BOTTOM‐UP PREDICTION OF INTER‐PATIENT VARIABILITY IN DRUG RESPONSE

13.4 PBPK/PD FOR PREDICTING THE INTER‐PATIENT VARIABILITY IN RESPONSE TO PRODRUGS AND ACTIVE METABOLITES

13.5 PBPK/PDWHEN SYSTEMIC CONCENTRATIONS ARE NOT THE DRIVER FOR DRUG RESPONSE

13.5.1 Pre‐Systemic Drug Target

13.5.2 Effect‐Site Drug Concentration Different from Systemic Concentration

13.6 PBPK/PD FOR MONOCLONAL ANTIBODIES

13.7 PBPK MODELS LINKED TO QUANTITATIVE SYSTEMS PHARMACOLOGY AND TOXICOLOGY MODELS

13.7.1 PBPK–QST Models to Predict Drug‐Induced Liver Injury

13.7.2 PBPK–QST Models to Predict Drug‐Induced Cardiotoxicity

13.8 CONCLUSIONS

REFERENCES

14 PBPK MODELING AND SIMULATIONS TO EVALUATE CLINICAL DRUG‐DRUG INTERACTIONS. CONTENTS

14.1 INTRODUCTION

14.2 CLINICAL DDI STUDIES AND MODELING APPROACHES TO ADDRESS KEY QUESTIONS RELATED TO DRUG–DRUG INTERACTIONS

14.2.1 Dedicated Clinical DDI Studies

14.2.2 Investigation of Phenotypic Effects for NMEs Predominantly Cleared by Polymorphic Enzyme or Transporter

14.2.3 Prospective Nested DDI Study

14.2.4 Cocktail DDI Study

14.2.5 PBPK Modeling and Simulations

14.2.6 Claims Relating to Results of DDI Studies

14.2.7 Impact on Label

14.3 PBPK MODELING OF DIFFERENT TYPES OF DRUG INTERACTIONS

14.3.1 PBPK Modeling Strategy: New Molecular Entity as Victim of CYP‐Based Drug Interactions

14.3.2 PBPK Modeling Strategy: New Molecular Entity as Perpetrator of CYP‐Based Drug Interactions

14.3.3 Non‐CYP Based Drug Interactions

14.3.4 Transporter‐Mediated Drug Interactions

14.4 DDI PREDICTIONS WITH PBPK MODELING AND SIMULATIONS IN CLINICAL DRUG DEVELOPMENT AND REGULATORY SUBMISSIONS. 14.4.1 DDI Predictions Along the Value Chain (Figure 14.5)

14.4.2 Possible Regulatory Outcomes, Based on the Predictions from a Verified and Validated PBPK Model

14.4.3 Regulatory Acceptance of PBPK Analyses Included in Regulatory Submissions

14.4.4 Predictive Performance of PBPK Models

14.5 COMPARISON OF DDI PREDICTION USING STATIC AND DYNAMIC MODELS

14.6 CONCLUSIONS

REFERENCES

15 DOSE EXTRAPOLATION ACROSS POPULATIONS (HEALTHY ADULT CAUCASIAN TO PEDIATRIC, PREGNANT WOMEN, DIFFERENT ETHNICITIES, GERIATRIC, SMOKERS AND OBESE POPULATIONS) CONTENTS

15.1 INTRODUCTION

15.2 PBPK MODELING STRATEGY FOR DOSE EXTRAPOLATION TO SPECIFIC POPULATIONS

15.3 POTENTIAL BENEFITS OF PBPK MODELING FOR DOSE EXTRAPOLATIONS TO SPECIFIC POPULATIONS

15.4 DOSE EXTRAPOLATIONS TO SPECIFIC POPULATIONS

15.4.1 Pediatric Starting Dose Selection

15.4.2 Pregnancy

15.4.3 Ethnicity – Japanese Population

15.4.4 Geriatric Population

15.4.5 Obese

15.4.6 Smokers

15.5 CONCLUSIONS

REFERENCES

16 DOSE EXTRAPOLATION ACROSS POPULATIONS: HEALTHY ADULT TO HEPATIC AND RENAL IMPAIRMENT POPULATIONS. CONTENTS

16.1 INTRODUCTION

16.2 PATHOPHYSIOLOGICAL CHANGES IN ORGAN IMPAIRMENT. 16.2.1 Hepatic Impairment

16.2.2 Renal Impairment

16.3 PBPK MODELING STRATEGY: MODEL DEVELOPMENT, VERIFICATION, VALIDATION, AND APPLICATION

16.4 BENEFITSOFAPPLYINGVALIDATEDPBPKMODELSTOORGAN‐IMPAIRED POPULATIONS

16.4.1 Enhancing Regulatory Confidence in the Application of PBPK Modeling for the Prediction of Exposure in the Organ‐Impaired Population

16.4.2 Contribution of PBPK to the Totality of Evidence in Evaluating the Effect of Renal Impairment on Drug Exposure to Inform Labelling

16.5 CONCLUSIONS

REFERENCES

17 ABSORPTION‐RELATED APPLICATIONS OF PBPK MODELING. CONTENTS

17.1 INTRODUCTION

17.2 IN VITRO – IN VIVO DISCONNECT, PARAMETER NON‐IDENTIFIABILITY AND THE IMPORTANCE OF IDENTIFYING FACTORS LIMITING ABSORPTION THROUGH A DECONVOLUTION OF THE MECHANISMS CONTRIBUTING TO GUT BIOAVAILABILITY

17.3 NON‐REGULATORY INTERNAL APPLICATIONS OF PBPK MODELING AND SIMULATIONS. 17.3.1 Prediction of Fraction Absorbed

17.3.2 Oral Formulation Development

17.4 REGULATORY APPLICATIONS OF PBPK MODELING AND SIMULATIONS

17.4.1 Food–Drug Interactions

17.4.1.1 Food Effect Predictions

17.4.1.2 PBPK Modeling Strategy for Food Effect Prediction

17.4.1.3 Evaluation of Food Effect Predictions

17.4.1.4 Benefits of Food Effect Predictions and Challenges

17.4.1.5 PBPK Predictions of Food Effect in the Future

17.4.2 Interactions of a Poorly Soluble Weak Base with Acid Reducing Agents (ARAs)

17.4.3 In Vitro – In Vivo Correlations (IVIVCs) to Serve as Surrogate for Bioequivalence Testing (Case Study 12)

17.4.4 Biowaivers Based on Virtual Bioequivalence

17.4.5 Virtual Bioequivalence of Locally Acting Products (LAPs)

17.5 CONCLUSIONS

REFERENCES

18 REGULATORY GUIDELINES ON THE REPORTING OF PHYSIOLOGICALLY BASED PHARMACOKINETIC (PBPK) MODELING ANALYSIS. CONTENTS

18.1 INTRODUCTION

18.2 FOOD AND DRUG ADMINISTRATION (FDA) GUIDELINES

18.3 EUROPEAN MEDICINES AGENCY (EMA) GUIDELINES

18.4 COMPARISON OF FDA AND EMA GUIDELINES

18.5 RISK‐INFORMED EVIDENTIARY FRAMEWORK TO ASSESS PBPK MODEL CREDIBILITY

18.6 DRUG MODEL VERIFICATION OF LOCALLY ACTING PRODUCTS (LAPs)

REFERENCES

19 RESOLVING THE CHALLENGES TO ESTABLISHING CONFIDENCE IN PBPK MODELS. CONTENTS

19.1 INTRODUCTION

19.2 REQUIREMENTS FOR DEVELOPING MECHANISTICALLY CREDIBLE PBPK MODELS FOR THE THREE BROAD CATEGORIES OF APPLICATIONS

19.3 CHALLENGES TO DEVELOPING MECHANISTICALLY CREDIBLE PBPK MODELS AND CONSEQUENCES

19.3.1 Model Building. 19.3.1.1 Identifying Key Mechanisms Impacting An Application

19.3.1.2 Model Parameterization with In Vitro Data in a Bottom‐Up Approach

19.3.1.3 Parameter Estimation from Clinical Data in Top‐Down or Middle‐Out Approaches

19.3.1.4 Parameter Non‐Identifiability as a Barrier to Deconvolute Mechanisms Contributing to Gut Bioavailability

19.3.2 Model Verification of Predicted Exposure and Validation of Predictive Performance

19.4 RESOLVING THE CHALLENGES TO DEVELOPING MECHANISTICALLY CREDIBLE PBPK MODELS

19.5 TOTALITY OF EVIDENCE

19.6 CONCLUSIONS

REFERENCES

20 EPILOGUE. CONTENTS

20.1 PBPK MODELING SUCCESSES

20.2 CHALLENGES

20.2.1 Drug Model Parameterization

20.2.2 Knowledge Gaps in Physiological Parameters

20.2.3 Prospective Validation of Prediction Performance

20.3 MEETING THE CHALLENGES

20.4 FUTURE DIRECTIONS FOR PBPK MODELING

REFERENCES

SECTION III. CASE STUDIES OF PBPK APPLICATIONS IN THE PHARMACEUTICAL INDUSTRY

CASE STUDY 1 HYPOTHESIS TESTING (SOLUBILITY) S1.1 IDENTIFICATION OF HIGHER IN VIVO SOLUBILITY THAN MEASURED IN VITRO. S1.1.1 Key Question

S1.1.2 Background

S1.1.3 Objective

S1.1.4 Preclinical Data Leading to Hypothesis Generation

S1.1.5 Data Needed for Initial Model Building

S1.1.6 Hypothesis Testing

S1.1.7 Further Verification of Hypothesis

S1.1.8 Learnings

REFERENCES

CASE STUDY 2 HYPOTHESIS TESTING (GASTRIC EMPTYING) S2.1 IDENTIFICATION OF GASTRIC EMPTYING-LIMITED ORAL DRUG ABSORPTION. S2.1.1 Key Question

S2.1.2 Background

S2.1.3 Objective

S2.1.4 Data Needed for Initial Model Building

S2.1.5 Hypothesis Testing

S2.1.6 Learnings

REFERENCES

CASE STUDY 3 HYPOTHESIS TESTING (INTESTINAL LOSS) S3.1 IDENTIFICATION OF INTESTINAL LOSS. S3.1.1 Key Question

S3.1.2 Background

S3.1.3 Objective

S3.1.4 Data Needed for Initial Model Building

S3.1.5 Hypothesis Testing

S3.1.6 Further Verification of Hypothesis

S3.1.7 Learnings

REFERENCES

CASE STUDY 4 PBPK/PD. Use of PBPK/PD for the Dose Optimization of the Antimicrobial Agent, Ciprofloxacin. S4.1 KEY QUESTION

S4.2 BACKGROUND

S4.3 OBJECTIVES

S4.4 DATA

S4.5 MODELING STRATEGY

S4.6 SENSITIVITY ANALYSIS

S4.7 CONCLUSION

REFERENCES

CASE STUDY 5 DRUG–DRUG INTERACTION (INHIBITION) Drug–Drug Interaction Risk Assessment for Midazolam Due to CYP3A Inhibition by Itraconazole. S5.1 KEY QUESTION

S5.2 BACKGROUND

S5.3 OBJECTIVES

S5.4 DATA

S5.5 MODELING STRATEGY

S5.6 SENSITIVITY ANALYSIS

S5.7 LEARNINGS

Additional questions that can be addressed with PBPK modelling

REFERENCES

CASE STUDY 6 DRUG–DRUG INTERACTION (INDUCTION) S6.1 DRUG–DRUG INTERACTION RISK ASSESSMENT FOR MIDAZOLAM DUE TO CYP3A INDUCTION BY RIFAMPICIN. S6.1.1 Key Question

S6.1.2 Background

S6.1.3 Objectives

S6.1.4 Data

S6.1.5 Modeling Strategy

S6.1.6 Sensitivity Analysis

REFERENCES

CASE STUDY 7 GENETIC POLYMORPHISM. S7.1 IMPACT OF GENETIC POLYMORPHISM ON THE PHARMACOKINETICS OF RISPERIDONE. S7.1.1 Key Question

S7.1.2 Background

S7.1.3 Objectives

S7.1.4 Modeling Strategy

S7.2 Results. S7.2.1 Sensitivity Analysis

REFERENCES

CASE STUDY 8 PEDIATRIC EXTRAPOLATION. S8.1 IMPACT OF UGT2B7 MATURATION ON THE PHARMACOKINETICS OF MORPHINE. S8.1.1 Key Question

S8.1.2 Background

S8.1.3 Objectives

S8.1.4 Modeling Strategy

S8.1.5 Morphine: Compound Data

S8.1.6 Morphine: Clinically Observed Data

S8.1.7 Sensitivity Analysis

S8.2 CONCLUSION

REFERENCES

CASE STUDY 9 PREGNANCY. S9.1 IMPACT OF PREGNANCY ON THE PHARMACOKINETICS OF METRONIDAZOLE. S9.1.1 Key Question

S9.1.2 Background

S9.1.3 Objectives

S9.1.4 Modeling Strategy

S9.2 RESULTS

REFERENCES

CASE STUDY 10 HEPATIC IMPAIRMENT. S10.1 IMPACT OF HEPATIC IMPAIRMENT ON THE PHARMACOKINETICS OF MIDAZOLAM AND LIDOCAINE. S10.1.1 Key Question

S10.1.2 Background

S10.1.3 Objectives

S10.2 MODELING STRATEGY

S10.3 RESULTS

REFERENCES

CASE STUDY 11 RENAL IMPAIRMENT. S11.1 IMPACT OF RENAL IMPAIRMENT ON THE PHARMACOKINETICS OF GENTAMICIN. S11.1.1 Key Question

S11.1.2 Background

S11.1.3 Objectives

S11.1.4 Modeling Strategy

S11.2 RESULTS

REFERENCES

CASE STUDY 12 ABSORPTION – IVIVC. S12.1 KEY QUESTION

S12.2 BACKGROUND

S12.3 OBJECTIVES

S12.4 DATA

S12.5 MODELING STRATEGY

S12.6 MODELING WORKFLOW

S12.7 SENSITIVITY ANALYSIS

S12.8 CONCLUSION

REFERENCES

APPENDICES. APPENDIX A: PHYSIOLOGICAL PARAMETERS IN PRECLINICAL SPECIES

Appendix B: Human

References

INDEX. A

B

C

D

E

F

G

H

I

K

L

M

N

O

P

Q

R

S

T

U

V

W

Y

Z

WILEY END USER LICENSE AGREEMENT

Отрывок из книги

Second Edition

.....

where fabs is the fraction of dose absorbed, fgut is the fraction escaping gut extraction, and fhep is the fraction escaping hepatic extraction. If the IV PK profile is available, knowing the hepatic clearance, hepatic extraction, EH, can be calculated using Equation 1.26. fhep is related to EH by

(1.37a)

.....

Добавление нового отзыва

Комментарий Поле, отмеченное звёздочкой  — обязательно к заполнению

Отзывы и комментарии читателей

Нет рецензий. Будьте первым, кто напишет рецензию на книгу Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulations
Подняться наверх