Drug Transporters

Drug Transporters
Автор книги: id книги: 2329531     Оценка: 0.0     Голосов: 0     Отзывы, комментарии: 0 25274,8 руб.     (275,65$) Читать книгу Купить и скачать книгу Купить бумажную книгу Электронная книга Жанр: Медицина Правообладатель и/или издательство: John Wiley & Sons Limited Дата добавления в каталог КнигаЛит: ISBN: 9781119739876 Скачать фрагмент в формате   fb2   fb2.zip Возрастное ограничение: 0+ Оглавление Отрывок из книги

Реклама. ООО «ЛитРес», ИНН: 7719571260.

Описание книги

DRUG TRANSPORTERS [b]Drug transporter fundamentals and relevant principles and techniques, featuring new and expanded chapters Drug Transporters: Molecular Characterization and Role in Drug Disposition provides in-depth analysis of the conceptual evolution and technical development for studying drug transporters. Contributions by an international panel of leading researchers address advances in transporters as drug targets, transporters in pharmacotherapy, the impact of transporters on drug efficacy and safety, the development of sophisticated model systems and sensitive assay methods, and more. Divided into two parts, the book first provides a thorough overview of relevant drug transporters before detailing the principles of drug transport and associated techniques. The updated and expanded third edition includes new chapters on in vitro-in vivo scale-up of drug transport activities, the ontogeny of drug transporters, the application of physiologically-based pharmacokinetic and pharmacodynamic modeling, and the use of transporters as therapeutic targets for diseases. Reflects the current state of the field and offers perspectives on future directions Covers basic knowledge, clinical outcomes, and emerging discoveries in transporter science Provides up-to-date information on drug transporter families, mechanisms, and clinical implications Includes extensive references and numerous figures and tables throughout Understandable for novices while offering sufficient depth for more experienced researchers, Drug Transporters: Molecular Characterization and Role in Drug Disposition, Third Edition is an excellent textbook for pharmacological or physiological science courses in drug/membrane transport, and an invaluable reference for academic or industrial scientists working in the transporter field and related areas of drug metabolism, pharmacokinetics, and pharmacodynamics.

Оглавление

Группа авторов. Drug Transporters

Table of Contents

List of Tables

List of Illustrations

Guide

Pages

DRUG TRANSPORTERS. Molecular Characterization and Role in Drug Disposition

LIST OF CONTRIBUTORS

PREFACE

1 OVERVIEW OF DRUG TRANSPORTER FAMILIES

1.1 INTRODUCTION

1.2 WHAT ARE DRUG TRANSPORTERS?

1.3 STRUCTURE AND MODEL OF DRUG TRANSPORTERS

1.4 TRANSPORT MECHANISMS

1.5 POLARIZED EXPRESSION OF DRUG TRANSPORTERS IN BARRIER EPITHELIUM

1.6 CLASSIFICATIONS OF DRUG TRANSPORTERS

1.6.1 Definition of Efflux and Influx Transporters

1.6.2 Definition of Absorptive and Secretory Transporters

1.6.3 Relationship Between Influx/Efflux and Absorptive/Secretory Transporters

1.6.4 ABC Transporters and SLC Transporters

1.7 NOMENCLATURE OF DRUG TRANSPORTERS

1.8 REGULATION OF DRUG TRANSPORTERS

1.9 CONCLUSION

REFERENCES

2 ORGANIC CATION AND ZWITTERION TRANSPORTERS

2.1 OVERALL INTRODUCTORY SECTION

CATION TRANSPORTERS: OCT1, OCT2, OCT3 (OTHER SECTION: SLC19A2, SLC19A3, PMAT) 2.2 INTRODUCTION TO THE OCT FAMILY

2.2.1 Tissue Distribution

2.2.2 Structure–Function Relationships

2.2.3 Transport Mechanism

2.3 OCT1. 2.3.1 Substrate and Inhibitor Selectivity

2.3.2 Regulation

2.3.3 Animal Models

2.3.4 Human Genetic Studies

2.3.5 Biomarkers and FDA Guidances for Transporter‐Mediated DDIs

2.4 OCT2. 2.4.1 Substrate and Inhibitor Selectivity

2.4.2 Regulation

2.4.3 Animal Models

2.4.4 Human Genetic Studies

2.5 OCT3. 2.5.1 Substrate and Inhibitor Selectivity

2.5.2 Regulation

2.5.3 Animal Models

2.5.4 Human Genetic Studies

2.6 OTHER IMPORTANT CATION TRANSPORTERS: SLC19A2, SLC19A3 (THIAMINE TRANSPORTERS), SLC29A4 (MONOAMINE TRANSPORTER) 2.6.1 Introduction

2.6.2 Tissue Distribution in Human

2.6.3 Substrate and Inhibitor Selectivity

2.6.4 Animal Models

2.6.5 Human Genetic Studies

ZWITTERION TRANSPORTERS: OCTN1, OCTN2, SLC22A15 AND SLC22A16 (OTHER SECTION: OCTN3) 2.7 INTRODUCTION TO THE ZWITTERION TRANSPORTERS

2.7.1 General Tissue Distribution

2.7.2 General Structure Function

2.7.3 Transport Mechanism

2.8 OCTN1. 2.8.1 Substrate and Inhibitor Selectivity

2.8.2 Regulation

2.8.3 Animal Models

2.8.4 Human Genetic Studies

2.9 OCTN2. 2.9.1 Substrate and Inhibitor Selectivity

2.9.2 Regulation

2.9.3 Animal Models

2.9.4 Human Genetic Studies

2.10 SLC22A15. 2.10.1 Introduction

2.10.2 Substrate and Inhibitor Selectivity

2.10.3 Human Genetic Studies

2.11 SLC22A16 (FLIPT2, CT2, OCT6) 2.11.1 Introduction

2.11.2 Substrate and Inhibitor Selectivity

2.11.3 Regulation

2.11.4 Human Genetic Studies

2.12 OCTN3. 2.12.1 Introduction

2.12.2 Substrate and Inhibitor Selectivity

2.12.3 Regulation

2.13 CONCLUSION

REFERENCES

3 MULTIDRUG AND TOXIN EXTRUSION PROTEINS

3.1 INTRODUCTION

3.2 TISSUE AND SUBCELLULAR DISTRIBUTION. 3.2.1 Tissue Distribution

3.2.2 Subcellular Localization

3.3 TRANSPORT ACTIVITY

3.3.1 Energetics of Transport

3.3.2 Substrates

3.3.2.1 Metformin

3.3.2.2 Platinum Drugs

3.3.2.3 Cardiovascular Drugs

3.3.2.4 Alkaloids

3.3.2.5 Paraquat

3.3.2.6 Endogenous Molecules

3.3.3 Inhibitors

3.4 STRUCTURE. 3.4.1 Modeling of Ligand Interactions

3.4.2 Secondary Structure

3.4.3 Structural Features

3.5 EXPRESSION AND REGULATION. 3.5.1 Transcriptional Regulation

3.5.2 Short‐Term Regulation

3.5.3 Sex and Age

3.5.4 Pregnancy

3.5.5 Disease Models. 3.5.5.1 Kidney Disease and Injury

3.5.5.2 Liver Disease and Injury

3.6 DRUG EFFICACY AND TOXICITY. 3.6.1 Clinical Substrates, Probes, and Inhibitors

3.6.2 Pharmacokinetic Drug Interactions

3.6.2.1 Metformin

3.6.2.2 Physiologically Based Pharmacokinetic Methods to Assess Interactions

3.6.2.3 Serum Creatinine and Kidney Function

3.6.2.4 Other Endogenous Probe SubstratesNMN has

3.7 PHARMACOGENETICS

3.7.1 Metformin Pharmacokinetics

3.7.2 Metformin Efficacy. 3.7.2.1 Diabetes

3.7.2.2 Nondiabetes Indications

3.7.3 Cisplatin Toxicity

3.7.4 Flutamide Liver Injury

3.8 CONCLUSIONS

ACKNOWLEDGMENT

REFERENCES

4 ORGANIC ANION TRANSPORTERS (OATs)

4.1 OAT FAMILY. 4.1.1 Introduction

4.1.2 Discovery

4.1.3 Nomenclature

4.2 MOLECULAR CHARACTERIZATION. 4.2.1 Genomics

4.2.2 Protein Structure

4.2.3 Mechanism of Substrate Translocation

4.3 EXPRESSION AND REGULATION OF OATS. 4.3.1 Tissue Distribution

4.3.2 Ontogeny

4.3.3 Transcriptional Regulation

4.3.4 Post‐translational Regulation

4.4 OAT SUBSTRATES AND DRUG–DRUG (DRUG–METABOLITE) INTERACTION. 4.4.1 Substrates

4.4.2 Substrate Specificity

4.4.3 Inhibitors

4.4.4 Natural Products and Herbal Medicines

4.4.5 Drug–Drug Interactions and Drug–Metabolite Interactions

4.5 SYSTEMS BIOLOGY OF OATS. 4.5.1 Physiological Role

4.5.2 Metabolic Pathways Regulated by OATs

4.5.3 Chronic Kidney Disease and Uremic Toxins

4.5.4 Pathophysiology

4.5.5 Clinical Pharmacology

4.5.6 Remote Sensing and Signaling Theory

4.6 CONCLUSIONS

ACKNOWLEDGMENTS

REFERENCES

5 ORGANIC ANION TRANSPORTING POLYPEPTIDES OATP

5.1 INTRODUCTION TO THE OATP SUPERFAMILY

5.1.1 Nomenclature

5.2 MOLECULAR CHARACTERISTICS OF OATPS

5.2.1 Protein Structure

5.2.2 Transport Mechanisms

5.3 EXPRESSION AND REGULATION OF OATPS. 5.3.1 Tissue Distribution

5.3.2 Ontogeny of Expression

5.3.3 Post‐Translational Regulation. 5.3.3.1 Phosphorylation

5.3.3.2 N‐Glycosylation

5.3.3.3 Adapter Protein Interactions

5.3.4 Transcriptional Regulation

5.3.4.1 Liver‐Enriched Transcription Factors

5.3.4.2 Farnesoid X Receptor

5.3.4.3 Liver X Receptor

5.3.4.4 Pregnane X Receptor

5.3.4.5 Vitamin D Receptor

5.3.4.6 Thyroid Hormone Receptor

5.3.4.7 Aryl Hydrocarbon Receptor

5.3.4.8 Hypoxia‐Inducible Factor 1

5.3.4.9 Wnt‐β‐Catenin Signaling

5.3.4.10 Inflammatory Cytokine Signaling

5.3.5 Epigenetic Regulation. 5.3.5.1 Promoter Methylation

5.3.5.2 Micro‐RNA Regulation

5.4 OATP SUBSTRATES, INHIBITORS, AND STIMULATORS. 5.4.1 Substrates

5.4.2 Inhibitors

5.4.3 Stimulators

5.5 PHARMACOLOGY OF OATPS. 5.5.1 Role of OATPs in Drug Disposition

5.5.2 In vitro and In vivo OATP Probe Substrates and Inhibitors

5.5.3 Endogenous Biomarkers of OATP Activity

5.5.4 PET, SPECT, and MR Imaging Markers

5.5.5 Drug, Food, and Formulation Interactions

5.5.6 Pharmacogenetics

5.5.6.1 SLCO1B1

5.5.6.2 SLCO1B3

5.5.6.3 SLCO1A2

5.5.6.4 SLCO2B1

5.5.7 Effects of Disease on OATP Expression and Activity

5.5.7.1 Liver Cirrhosis

5.5.7.2 Nonalcoholic Fatty Liver Disease

5.5.7.3 Cholestatic Liver Disease

5.5.7.4 Chronic Kidney Disease

5.5.7.5 Inflammatory Diseases

5.6 PHYSIOLOGY AND PATHOPHYSIOLOGY OF OATPS. 5.6.1 Bilirubin Homeostasis

5.6.2 Bile Acid Homeostasis

5.6.3 Thyroid Hormones Homeostasis

5.6.4 Prostaglandins Homeostasis

5.6.5 Steroid Hormone Homeostasis

5.6.6 Cancers

5.6.6.1 Breast Cancer

5.6.6.2 Gastrointestinal Cancers

5.6.6.3 Prostate Cancer

5.6.7 Cardiovascular Disease

5.7 CONCLUSIONS

ACKNOWLEDGMENTS

REFERENCES

6 MAMMALIAN OLIGOPEPTIDE TRANSPORTERS

6.1 INTRODUCTION

6.1.1 Peptide and Protein Permeation Routes Across the GI Epithelium

6.2 OLIGOPEPTIDE TRANSPORTERS

6.2.1 Molecular and Structural Characteristics of Oligopeptide Transporters

6.3 FUNCTIONAL PROPERTIES. 6.3.1 Mechanism of Transport

6.3.2 Molecular Requirements for Substrate Recognition and Transport

6.3.3 General Substrate Specificities

6.3.4 Established Endogenous and Exogenous Substrates

6.4 REGULATION

6.4.1 Dietary Regulation

6.4.2 Developmental Regulation

6.4.3 Regulation by Circadian Rhythms

6.4.4 Disease State‐Dependent Regulation

6.4.5 Hormonal Regulation

6.4.6 Regulation by Pharmaceutical Agents

6.4.7 Single Nucleotide Polymorphisms

6.4.8 Splice Variants

6.5 PHARMACEUTICAL DRUG SCREENING

6.5.1 Case Study: Targeting Peptide Transporters for Increased Oral Absorption: Valacyclovir

6.6 CONCLUDING REMARKS

REFERENCES

7 MONOCARBOXYLIC ACID TRANSPORTERS

7.1 INTRODUCTION

7.2 SLC5 TRANSPORTER FAMILY. 7.2.1 Structure, Function, Localization, and Substrates. 7.2.1.1 Structure

7.2.1.2 Localization and Function

7.2.2 Regulation

7.2.3 Role in Health and Disease

7.2.4 Role in Drug Disposition

7.3 SLC16 TRANSPORTER FAMILY. 7.3.1 Structure, Function, Localization, and Substrates. 7.3.1.1 Structure and Function

7.3.1.2 Localization and Substrates

7.3.2 Regulation. 7.3.2.1 Transcriptional Regulation

7.3.2.2 Epigenetic Regulation

7.3.2.3 Post‐Transcriptional Regulation

7.3.2.4 Membrane Trafficking Regulation

7.3.2.5 Regulation of Transporter Activity

7.3.3 Role in Health and Disease. 7.3.3.1 Monocarboxylate Transporters 1, 2, and 4 (SLC16A1, SLC16A7, SLC16A3)

7.3.3.2 Monocarboxylate Transporter 3 (SLC16A8)

7.3.3.3 Monocarboxylate Transporter 5 (SLC16A4)

7.3.3.4 Monocarboxylate Transporter 6 (SLC16A5)

7.3.3.5 Monocarboxylate Transporter 7 (SLC16A6)

7.3.3.6 Monocarboxylate Transporter 8 (SLC16A2)

7.3.3.7 Monocarboxylate Transporter 9 (SLC16A9)

7.3.3.8 Monocarboxylate Transporter 10 (SLC16A10)

7.3.3.9 Monocarboxylate Transporter 11 (SLC16A11)

7.3.3.10 Monocarboxylate Transporter 12 (SLC16A12)

7.3.3.11 Monocarboxylate Transporter 13/14 (SLC16A13/SLC16A14)

7.3.4 Role in Drug Disposition

7.3.4.1 Role of MCTs in the Disposition of GHB

7.3.4.2 Role of MCTs in the Disposition of Other Drugs

7.4 CONCLUSIONS AND FUTURE PERSPECTIVES

REFERENCES

8 THE NUCLEOSIDE TRANSPORTERS CNTs AND ENTs

8.1 INTRODUCTION

8.2 MOLECULAR AND FUNCTIONAL CHARACTERISTICS OF CNTs (SLC28) 8.2.1 Family Members and Substrate Specificity of CNTs

8.2.2 Transport Mode of CNTs

8.2.3 Tissue Distribution and Cellular Localization of CNTs

8.2.4 Interaction with Nucleoside Analogs

8.2.5 Structure–Function Relationship of CNTs

8.3 MOLECULAR AND FUNCTIONAL CHARACTERISTICS OF ENTs (SLC29) 8.3.1 Family Members and Substrate Specificity of ENTs

8.3.2 Transport Mode of ENTs

8.3.3 Tissue Distribution and Cellular Localization of ENTs

8.3.4 Interaction with Nucleoside Analogs

8.3.5 Structure–Function Relationship of ENTs

8.4 METHODS AND MOUSE MODELS TO STUDY ENTs AND CNTs

8.5 REGULATION OF CNTs AND ENTs

8.6 PHYSIOLOGICAL AND PATHOPHYSIOLOGICAL FUNCTIONS OF CNTs AND ENTs. 8.6.1 Nucleoside Homeostasis

8.6.2 Adenosine Signaling

8.6.3 ENTs and CNTs in AMPK Signaling

8.6.4 ENT1 and ENT3 in Hematopoiesis/Erythropoiesis

8.6.5 ENT3 in Autosomal Recessive Disorders

8.6.6 CNT1 in Inherited Metabolic Disorders

8.7 THERAPEUTIC SIGNIFICANCE OF CNTs AND ENTs

8.7.1 CNTs and ENTs in Intracellular Disposition of Nucleoside Drugs

8.7.2 CNTs and ENTs in Pharmacokinetics of Nucleoside Drugs

8.7.3 CNTs and ENTs as Drug Targets

8.8 CONCLUSIONS

ACKNOWLEDGMENT

REFERENCES

9 BILE ACID TRANSPORTERS

9.1 INTRODUCTION

9.2 OVERVIEW OF TRANSPORTERS IN THE ENTEROHEPATIC CIRCULATION OF BILE ACIDS

9.3 HEPATOBILIARY BILE ACID TRANSPORTERS. 9.3.1 BSEP/ABCB11: Bile Salt Export Pump

9.3.2 NTCP/SLC10A1: Sodium Taurocholate Cotransporting Polypeptide

9.4 INTESTINAL BILE ACID TRANSPORTERS. 9.4.1 ASBT/SLC10A2: Apical Sodium‐Dependent Bile Acid Transporter

9.4.2 OSTα‐OSTβ/SLC51A/SLC51B: The Basolateral Heteromeric Organic Solute Transporter

9.5 CONCLUSIONS AND FUTURE PERSPECTIVES

REFERENCES

10 THE P‐GLYCOPROTEIN MULTIDRUG TRANSPORTER

10.1 MULTIDRUG RESISTANCE AND P‐GLYCOPROTEIN

10.2 EXPRESSION PATTERN OF P‐GLYCOPROTEIN

10.3 SUBSTRATES AND INHIBITORS OF P‐GLYCOPROTEIN

10.4 STRUCTURE OF P‐GLYCOPROTEIN

10.5 EPITOPE MAPPING OF MONOCLONAL ANTIBODIES SPECIFIC FOR HUMAN P‐GLYCOPROTEIN (MRK‐16, 4E3, AND UIC2)

10.6 MOLECULAR MECHANISM OF POLYSPECIFICITY

10.7 DRUG TRANSPORT CYCLE OF P‐GLYCOPROTEIN

10.8 EFFORTS TO DISCOVER MODULATORS

10.9 ROLE OF P‐GLYCOPROTEIN IN THE PHYSIOLOGY AND BIOAVAILABILITY OF DRUGS

10.10 CONCLUSIONS AND FUTURE PERSPECTIVE

ACKNOWLEDGMENTS

REFERENCES

11 MULTIDRUG RESISTANCE PROTEINS OF THE ABCC SUBFAMILY

11.1 INTRODUCTION

11.2 NOMENCLATURE AND MOLECULAR CHARACTERIZATION

11.3 EXPRESSION AND LOCALIZATION OF ABCC TRANSPORTERS IN NORMAL HUMAN TISSUES AND IN HUMAN CANCERS

11.4 FUNCTIONAL PROPERTIES/SUBSTRATE SPECIFICITY AND MULTIDRUG RESISTANCE PROFILES OF HUMAN ABCC/MRPS

11.5 CLINICAL CONSEQUENCES OF GENETIC VARIANTS IN ABCC GENES

11.5.1 Genetic Variants of Human ABCC/MRP Genes and the Mendelian Inheritance of Diseases and Syndromes

11.5.1.1 ABCC2/MRP2 and Dubin–Johnson Syndrome

11.5.1.2 ABCC6/MRP6 and Pseudoxanthoma Elasticum

11.5.1.3 ABCC11/MRP8 and Earwax type

11.5.2 Genetic Variants of Human ABCC/MRP Genes and Clinical Consequences on Drug Response and Susceptibility to Complex Disease. 11.5.2.1 ABCC1/MRP1

11.5.2.2 ABCC2/MRP2

11.5.2.3 ABCC3/MRP3

11.5.2.4 ABCC4/MRP4

11.5.2.5 ABCC5/MRP5

11.5.2.6 ABCC6/MRP6

11.5.2.7 ABCC10/MRP7

11.5.2.8 ABCC11/MRP8

11.6 CONCLUSION

ACKNOWLEDGMENTS

REFERENCES

12 ABCG2, THE BREAST CANCER RESISTANCE PROTEIN (BCRP)

12.1 DISCOVERY AND NOMENCLATURE

12.2 THE ABCG2 GENE AND EXPRESSION. 12.2.1 The ABCG2 Gene

12.2.2 Factors Controlling ABCG2 Expression

12.3 PHYSICAL PROPERTIES. 12.3.1 Structure

12.3.2 Trafficking and Regulation of Cell Surface Expression

12.4 SUBSTRATES AND INHIBITORS OF BCRP. 12.4.1 Endogenous Substrates

12.4.2 Exogenous Substrates

12.4.3 Inhibitors

12.5 RECENT FINDINGS CONCERNING PHYSIOLOGICAL FUNCTIONS. 12.5.1 ABCG2, Urate, and Gout

12.5.2 Jr(a‐) Phenotype

12.6 PREDICTED PHYSIOLOGICAL FUNCTION FROM TISSUE DISTRIBUTION

12.6.1 Stem Cells

12.6.2 Placenta

12.6.3 Mammary Gland

12.6.4 Testis

12.6.5 Blood–Brain Barrier

12.6.6 Liver and the Gastrointestinal Tract

12.6.7 Kidneys

12.7 ABCG2 EXPRESSION IN CANCER AND ITS ROLE IN DRUG RESISTANCE

12.8 GENETIC POLYMORPHISMS

12.8.1 Genetic Variants of ABCG2

12.8.2 The Q141K (rs2231142) SNP and Drug Disposition/Clinical Outcome

12.8.3 Other ABCG2 Polymorphisms and Drug Disposition/Clinical Outcome

12.9 CONCLUSION

REFERENCES

13 DRUG TRANSPORT IN THE LIVER

13.1 INTRODUCTION

13.2 HEPATIC PHYSIOLOGY: LIVER STRUCTURE AND FUNCTION

13.3 HEPATIC TRANSPORT PROTEINS

13.3.1 Uptake Transporters

13.3.2 Canalicular (Apical) Efflux Transporters

13.3.3 Basolateral Efflux Transporters

13.4 REGULATION OF HEPATIC TRANSPORT PROTEINS IN HUMANS

13.4.1 Transcriptional Regulation

13.4.2 Post‐Translational Regulation

13.4.3 Transporter Induction

13.5 PHYSIOLOGICAL FACTORS THAT INFLUENCE HEPATIC DRUG TRANSPORT PROTEINS IN HUMANS. 13.5.1 Ontogeny and Effects of Aging on Hepatic Transporters

13.5.2 Microbiome Effects on Hepatic Transporters

13.6 DISEASE‐RELATED ALTERATIONS IN HUMAN HEPATIC TRANSPORT PROTEINS

13.6.1 Hepatic Transporter‐Mediated Liver Diseases

13.6.1.1 Cholestatic Disorders

13.6.1.2 Dubin–Johnson Syndrome

13.6.1.3 Rotor Syndrome

13.6.2 Liver Disease‐Mediated Alterations in Hepatic Transporters

13.6.2.1 Cholestasis

13.6.2.2 Nonalcoholic Fatty Liver Disease

13.6.2.3 Cirrhosis

13.6.2.4 Hepatocellular Carcinoma

13.6.2.5 Autoimmune Hepatitis

13.6.2.6 Alcoholic Liver Disease

13.6.2.7 Chronic Hepatitis C Viral Infection

13.6.3 Other Non‐Hepatic Disease‐Mediated Alterations in Hepatic Transporters. 13.6.3.1 Renal Diseases

13.6.3.2 Sepsis/Acute Inflammation

13.7 METHODS FOR STUDYING HEPATOBILIARY DRUG TRANSPORT

13.7.1 In vitro Systems

13.7.2 In vivo Systems

13.7.3 In silico Approaches

13.8 HEPATIC TRANSPORTER‐MEDIATED DRUG–DRUG INTERACTIONS (DDIS)

13.9 INTERPLAY BETWEEN DRUG METABOLISM AND TRANSPORT

13.10 HEPATIC TRANSPORT PROTEINS AS DETERMINANTS OF DRUG TOXICITY

13.11 THE FUTURE OF HEPATIC DRUG TRANSPORT

ACKNOWLEDGMENTS

REFERENCES

14 DRUG TRANSPORT IN THE BRAIN

14.1 INTRODUCTION

14.2 PHYSIOLOGY OF THE BRAIN BARRIERS AND BRAIN PARENCHYMA. 14.2.1 Blood–Brain Barrier/Neurovascular Unit

14.2.1.1 Endothelial Cells

14.2.1.2 Pericytes

14.2.1.3 Astrocytes

14.2.1.4 Neurons

14.2.1.5 Other Glial Cells in the Brain Parenchyma (Oligodendrocytes, Microglia)

14.2.2 Blood–Cerebrospinal Fluid Barrier

14.2.3 Blood–Arachnoid Barrier

14.3 FUNCTIONAL EXPRESSION AND LOCALIZATION OF DRUG TRANSPORTERS IN THE BRAIN. 14.3.1 ABC Transporters

14.3.1.1 P‐glycoprotein (P‐gp)

14.3.1.2 Breast Cancer Resistance Protein (BCRP/Bcrp)

14.3.1.3 Multidrug Resistance Proteins (MRPs/Mrps)

14.3.2 Solute Carrier (SLC) Transporter Family

14.3.2.1 Organic Cation Transporters (OCTs/Octs) and Novel Organic Cation Transporters (OCTNs/Octns)

14.3.2.2 Organic Anion Transporters (OATs)

14.3.2.3 Organic Anion Transporting Polypeptides (OATPs/Oatps)

14.3.2.3.1 Human OATP Isoforms

14.3.2.3.2 Rodent Oatp Isoforms

14.3.2.4 Peptide Transporters

14.3.2.5 Nucleoside Transporters. 14.3.2.5.1 Concentrative Nucleoside Transporters (CNTs)

14.3.2.5.2 Equilibrative Nucleoside Transporters (ENTs)

14.3.2.6 Folate Receptors/Transporters

14.3.2.6.1 Folate Receptors

14.3.2.6.2 Reduced Folate Carrier

14.3.2.6.3 Proton‐Coupled Folate Transporter

14.4 RELEVANCE OF DRUG TRANSPORTERS IN CNS DISORDERS. 14.4.1 HIV‐1 Infection of the Brain

14.4.2 Neurodegenerative Disorders. 14.4.2.1 Parkinson’s Disease

14.4.2.2 Alzheimer’s Disease

14.4.2.3 Multiple Sclerosis

14.4.3 Epilepsy

14.4.4 Brain Neoplasia

14.4.5 Cerebral Hypoxia and Ischemic Stroke

14.4.6 Traumatic Brain Injury

14.4.7 Pain

14.4.8 Cerebral Folate Deficiency Disorders

14.5 CONCLUSION

REFERENCES

15 DRUG TRANSPORT IN THE KIDNEY

15.1 KIDNEY STRUCTURE AND FUNCTION

15.2 MAJOR DRUG TRANSPORTERS EXPRESSED IN HUMAN KIDNEY

15.3 TARGETED PROTEOMICS OF HUMAN RENAL TRANSPORTERS

15.4 HUMAN TRANSPORTERS IN RENAL INJURY AND DISEASE. 15.4.1 Ischemia/Reperfusion Injury

15.4.2 Glucose Transporters and Diabetes

15.4.3 Chronic Kidney Disease

15.4.4 Hyperuricemia and Gout

15.5 CONCLUSIONS AND FUTURE PERSPECTIVES

REFERENCES

16 DRUG TRANSPORTERS IN THE HUMAN INTESTINE

16.1 INTRODUCTION

16.2 INTESTINAL ABC TRANSPORTERS (EXPRESSION, LOCALIZATION, FUNCTION)

16.3 INTESTINAL SLC TRANSPORTERS (EXPRESSION, LOCALIZATION, FUNCTION)

16.4 INTERPLAY OF APICAL AND BASOLATERAL TRANSPORTERS

16.5 VARIABILITY OF INTESTINAL TRANSPORTERS

16.5.1 Transcriptional Regulation by Nuclear Receptors

16.5.2 Epigenetic Regulation

16.5.3 Disease‐Related Changes

16.6 DRUG–DRUG INTERACTIONS INVOLVING INTESTINAL TRANSPORTERS

16.7 LIMITATIONS OF AVAILABLE RESEARCH MODELS FOR THE HUMAN INTESTINE

16.8 CONCLUSIONS AND FUTURE PERSPECTIVES

REFERENCES

17 DRUG TRANSPORT IN THE PLACENTA

17.1 INTRODUCTION

17.2 BLOOD–PLACENTAL BARRIER RELEVANT TO DRUG PERMEABILITY AND TRANSPORT

17.3 DRUG TRANSPORTERS IN HUMAN PLACENTA

17.3.1 ABC Transporters in Human Placenta. 17.3.1.1 P‐glycoprotein (P‐gp, MDR1, or ABCB1)

17.3.1.2 Breast Cancer Resistance Protein (BCRP or ABCG2)

17.3.1.3 Multidrug Resistance Proteins (MRPs or ABCCs)

17.3.2 SLC Transporters in Human Placenta. 17.3.2.1 Monoamine Transporters

17.3.2.2 Organic Cation and Anion Transporters

17.3.2.3 Organic Anion Transporting Polypeptides (OATPs)

17.3.2.4 Monocarboxylate Transporters MCT1 (SLC16A1) and MCT4 (SLC16A3)

17.3.2.5 Folate Transporters RFT1 (SLC19A1) and PCFT (SLC46A1)

17.3.2.6 Equilibrative Nucleoside Transporters

17.4 METHODS TO STUDY PLACENTAL DRUG TRANSPORT

17.5 CONCLUSIONS AND FUTURE PERSPECTIVES

REFERENCES

18 POLYMORPHISMS OF DRUG TRANSPORTERS AND CLINICAL RELEVANCE

18.1 INTRODUCTION

18.2 GENETIC VARIATION IN DRUG TRANSPORTERS LEADING TO ALTERED EFFECT

18.2.1 ABCB1 (MDR1, P‐Glycoprotein)

18.2.2 ABCG2 (BCRP, Breast Cancer Resistance Protein)

18.2.3 SLCO1B1 (OATP1B1, Organic Anion Transporter Polypeptide 1B1)

18.2.4 SLC22A1 (OCT1, Organic Cation Transporter 1)

18.2.5 SLC22A2 (OCT2, Organic Cation Transporter 2)

18.2.6 SLC47A1/SLC47A2 (MATE1/MATE2‐K, Multidrug and Toxin Extrusion Protein 1/2‐K)

18.2.7 SLC22A6/SLC22A8 (OAT1/OAT3, Organic Anion Transporter 1/3)

18.3 DISCUSSION

18.4 PERSPECTIVES

REFERENCES

19 ONTOGENY OF DRUG TRANSPORTERS

19.1 INTRODUCTION

19.1.1 Pediatric Populations

19.1.2 Assessment of Ontogeny

19.2 TRANSPORTER ONTOGENY

19.2.1 Liver. 19.2.1.1 ABC Transporters. 19.2.1.1.1 Breast Cancer Resistance Protein (BCRP)

19.2.1.1.2 Bile Salt Export Pump (BSEP)

19.2.1.1.3 Multidrug Resistance Protein (MRP)

19.2.1.1.4 P‐Glycoprotein (PGP)

19.2.1.2 SLC Transporters. 19.2.1.2.1 Multidrug and Toxin Extrusion (MATE)

19.2.1.2.2 Monocarboxylate Transporters (MCTs)

19.2.1.2.3 Organic Anion Transporters (OATs)

19.2.1.2.4 Organic Anion Transporting Polypeptides (OATPs)

19.2.1.2.5 Organic Cation Transporters (OCTs)

19.2.1.2.6 Organic Cation Transporter Novel (OCTN)

19.2.1.2.7 Organic Solute Transporter (OST)

19.2.1.2.8 Sodium/Taurocholate Cotransporting Polypeptide (NTCP)

19.2.2 Intestine. 19.2.2.1 ABC Transporters. 19.2.2.1.1 BCRP

19.2.2.1.2 MRPs

19.2.2.1.3 PGP

19.2.2.2 SLC Transporters. 19.2.2.2.1 Apical Sodium Dependent Bile Acid Transporter (ASBT)

19.2.2.2.2 OATP

19.2.2.2.3 OCT

19.2.2.2.4 OSTα/β

19.2.2.2.5 Peptide Transporters (PEPT)

19.2.3 Kidney. 19.2.3.1 ABC Transporters. 19.2.3.1.1 BCRP

19.2.3.1.2 MRPs

19.2.3.1.3 PGP

19.2.3.2 SLC Transporters. 19.2.3.2.1 MATE

19.2.3.2.2 OATs

19.2.3.2.3 OATPs

19.2.3.2.4 OCTs

19.2.3.2.5 OCTNs

19.2.3.2.6 PEPT

19.3 CHALLENGES IN ASSESSING TRANSPORTER ONTOGENY

19.4 CONCLUSIONS AND FUTURE PERSPECTIVES

REFERENCES

20 EXPERIMENTAL APPROACHES FOR STUDYING DRUG TRANSPORTERS

20.1 INTRODUCTION

20.2 GENETICALLY MODIFIED AND HUMAN‐XENOGRAFT MODEL ANIMALS. 20.2.1 Gene Knockout Rodents

20.2.2 Transient Knockdown Rodents

20.2.3 Genetically Humanized Mice

20.2.4 Human Organ Xenograft Mice

20.3 IN VIVO EXPERIMENTS

20.3.1 Pharmacokinetic Analysis

20.3.2 Integration Plot Analysis

20.3.3 Imaging Analysis

20.3.4 Metabolome Analysis

20.3.5 Microdialysis

20.4 ISOLATED TISSUE METHODS

20.4.1 Transport Studies in Intestinal Tissues. 20.4.1.1 The Loop Method

20.4.1.2 The Ussing‐Type Chamber Method

20.4.1.3 The Everted Sac Method

20.4.2 Sliced Organs

20.4.3 Perfused Organs

20.5 PRIMARY CELL CULTURES, ESTABLISHED MODEL CELL LINES, AND THEIR COMBINATION

20.5.1 Cryopreserved and Freshly Isolated Hepatocytes

20.5.2 Isolated Renal Proximal Tubule Cells

20.5.3 Established Cell Lines

20.5.4 Conditionally Immortalized Cell Lines

20.5.5 iPS Cells

20.5.6 Heterologous Expression of Drug Transporters in Immortalized Cell Lines

20.5.7 Heterologous Expression of Drug Transporters in Xenopus laevis Oocytes

20.5.8 Microphysiological System

20.6 ANALYSIS OF THE TRANSPORT, INHIBITION, INDUCTION, AND DOWNREGULATION OF TRANSPORTERS

20.6.1 Uptake Studies in Cultured Cells

20.6.2 Transcellular Transport Studies

20.6.3 Analysis of the Direct Inhibition of Transporters

20.6.4 Assay of Preincubation‐Dependent and Long‐Lasting Inhibition

20.6.5 Induction and Downregulation of Transporters

20.6.6 Quantification of Transporter Protein Expression

20.6.7 Screening of Transporters Based on a Genetic Approach

20.7 MEMBRANE VESICLES

20.7.1 Preparation of Membrane Vesicles from Tissues

20.7.2 Preparation of Membrane Vesicles from Gene Expression Systems

20.7.3 Purification and Reconstitution of Transporters

20.7.4 Methods in Transport Studies Using Membrane Vesicles

20.8 PERSPECTIVES

REFERENCES

21 TRANSPORTERS‐MEDIATED DRUG DISPOSITION—PHYSIOCHEMISTRY AND IN SILICO APPROACHES

21.1 INTRODUCTION

21.2 PHYSICOCHEMICAL DETERMINANTS OF HEPATOBILIARY TRANSPORT

21.3 IN SILICO MODELS FOR BILIARY ELIMINATION

21.4 PHYSICOCHEMICAL DETERMINANTS OF RENAL ELIMINATION

21.5 IN SILICO MODELS FOR RENAL ELIMINATION OR RENAL CLEARANCE

21.6 FRAMEWORK TO PREDICT TRANSPORTER‐MEDIATED CLEARANCE MECHANISM—EXTENDED CLEARANCE CLASSIFICATION SYSTEM (ECCS)

21.7 IN SILICO APPROACHES AND SAR OF CLINICALLY RELEVANT TRANSPORTERS. 21.7.1 P‐glycoprotein (P‐gp)

21.7.2 Multidrug Resistance Associated Protein 2 (MRP2)

21.7.3 Breast Cancer Resistant Protein (BCRP)

21.7.4 Organic Anion Transporting Polypeptide Transporters (OATPs)

21.7.5 Organic Anion Transporters (OATs) and Organic Cation Transporters (OCTs)

21.8 STRATEGIES TO ASSESS TRANSPORTER INVOLVEMENT DURING DRUG DISCOVERY

21.9 TARGETING DRUG TRANSPORTERS: CASE EXAMPLES. 21.9.1 Design of Hepatoselective Agent—Targeting OATP1B1/1B3

21.9.2 Intestine‐Specific P‐glycoprotein Inhibitor

21.10 CONCLUSIONS

REFERENCES

22 IN VITRO–IN VIVO SCALE‐UP OF DRUG TRANSPORT ACTIVITIES

22.1 INTRODUCTION

22.2 THEORETICAL BACKGROUND FOR THE PREDICTION OF IN VIVO PHARMACOKINETICS FROM IN VITRO DATA

22.2.1 Contribution of Each Transporter to the Overall Membrane Transport

22.2.2 Analysis of the Rate‐Determining Process in the Hepatic Elimination Clearance of Drugs Based on the Extended Clearance Concept

22.2.3 Relationship Between the Overall Intrinsic Organ Clearance and Organ Clearance

22.3 PREDICTION OF HEPATIC TRANSPORT FROM IN VITRO DATA. 22.3.1 Prediction of Hepatic Uptake Clearance from In Vitro Experiments with Hepatocytes

22.3.2 “Albumin‐Mediated” Hepatic Uptake Mechanism: Importance in the In Vitro–In Vivo Extrapolation of Hepatic Uptake Clearance

22.3.3 Prediction of Biliary Excretion Clearance

22.4 PREDICTION OF RENAL TRANSPORT FROM IN VITRO DATA

22.5 PREDICTION OF TRANSPORTER‐MEDIATED DRUG–DRUG INTERACTIONS FROM IN VITRO DATA. 22.5.1 Background Theory

22.5.2 Possible Cause for the Discrepancy of In Vitro and In Vivo Kinetic Parameters Used for the Estimation of DDI Risks

22.5.3 Prediction of Complex DDIs by In Vitro Data and PBPK Models Utilizing the “Middle‐Out” Approach

22.6 CONCLUSIONS AND PERSPECTIVES

REFERENCES

23 APPLICATION OF PHYSIOLOGICALLY BASED PHARMACOKINETIC AND PHARMACODYNAMIC (PBPK/PD) MODELING COMPRISING TRANSPORTERS: DELINEATING THE ROLE OF VARIOUS FACTORS IN DRUG DISPOSITION AND TOXICITY

23.1 INTRODUCTION ‐ THE RISE OF PHYSIOLOGICALLY‐BASED PHARMACOKINETIC AND PHARMACODYNAMIC (PBPK/PD) APPLICATIONS IN INDUSTRY, ACADEMIA AND REGULATORY SPACE

23.2 MODELS AND THEIR ASSUMPTIONS

23.2.1 PBPK Model Principles (A focused appraisal of core principles related to PBPK models that include transporters)

23.2.2 Modeling Permeability‐Limited Organ Models

23.2.2.1 General Concepts and Considerations of Permeability‐Limited Organ Modeling

23.2.2.2 Permeability‐Limited Liver Modeling

23.2.2.3 Permeability‐Limited Kidney Modeling

23.2.2.4 Permeability‐Limited Models for the Intestine

23.2.2.5 Permeability‐Limited Brain Models

23.2.2.6 Other Organs for Consideration for Permeability‐Limited Application

23.3 REAL‐WORLD APPLICATIONS OF TRANSPORTER ACTIVITY IN PBPK MODELS

23.3.1 Application I—Simultaneous Assessment of Transporters and Enzymes Impacting on Oral Bioavailability Using PBPK

23.3.2 Application II—Factoring in Metabolites When Considering Simultaneous Transporter‐Mediated DDIs

23.3.3 Application III—Addressing Population Variability for Combined Enzyme and Transporter‐Mediated Disposition: Application of Models for Disease Populations

23.3.4 Application IV—Extending Transporter Models—A Case for Applying Electrochemical Gradient Approach to Address Transporter Functionality

23.3.5 Application V—Toxicity

23.3.6 Application VI—Pharmacodynamics (Including Untangling the Impact of Transporters on the Variability of Pharmacokinetics versus Pharmacodynamics)

23.4 CONCLUSIONS AND FUTURE PERSPECTIVES

REFERENCES

24 TRANSPORTERS AS THERAPEUTIC TARGETS IN HUMAN DISEASES

24.1 INTRODUCTION

24.2 SLC TRANSPORTERS IN DISEASE AS TARGETS AND MODULATORS. 24.2.1 SLC Transporters as Targets in Cancer

24.2.1.1 Targeting Transporters with Imaging Agents in Cancer

24.2.2 SLC Transporters as Targets in Neurological Diseases

24.2.2.1 Major Depressive Disorder

24.2.2.2 Amyotrophic Lateral Sclerosis

24.2.2.3 Schizophrenia

24.2.2.4 Pain

24.2.3 SLC Transporters as Targets in Cardiovascular Disease, Hypertension, and Metabolic Diseases. 24.2.3.1 Cardiovascular Disease and Hypertension

24.2.3.2 Metabolic Diseases

24.3 ABC TRANSPORTERS IN DISEASE AS TARGETS AND MODULATORS. 24.3.1 ABC Transporters in Cancer as Modulators and Targets

24.3.2 ABC Transporters as Targets in Neurological Diseases

24.3.2.1 Alzheimer’s Disease

24.3.2.2 X‐Linked Adrenoleukodystrophy (X‐ALD)

24.4 CONCLUSIONS AND FUTURE PERSPECTIVES

ACKNOWLEDGMENTS

REFERENCES

25 DIET/NUTRIENT INTERACTIONS WITH DRUG TRANSPORTERS

25.1 INTRODUCTION

25.2 DIET/NUTRIENT INTERACTIONS WITH DRUG TRANSPORTERS. 25.2.1 Interactions of Diet/Dietary Supplements with Drug Transporters

25.2.1.1 St. John’s Wort

25.2.1.1.1 Effects on Drug Transporters

25.2.1.1.2 Clinical Drug Interactions

25.2.1.2 Grapefruit Juice

25.2.1.2.1 Effects on Drug Transporters

25.2.1.2.2 Clinical Drug Interactions

25.2.1.3 Green Tea

25.2.1.3.1 Effects on Drug Transporters

25.2.1.3.2 Clinical Drug Interactions

25.2.1.4 Ginseng

25.2.1.5 Turmeric

25.2.1.6 Red Wine

25.2.1.7 Ginkgo

25.2.2 Interactions of Flavonoids with Drug Transporters

25.2.2.1 Interactions with P‐gp

25.2.2.2 Interactions with MRPs

25.2.2.3 Interactions with BCRP

25.2.2.4 Interactions with OATP

25.2.2.5 Interactions with OAT

25.2.3 Interactions of Isothiocyanates with Drug Transporters

25.3 CONCLUSIONS

ACKNOWLEDGMENTS

REFERENCES

INDEX

Wiley Series in Drug Discovery and Development

WILEY END USER LICENSE AGREEMENT

Отрывок из книги

Wiley Series in Drug Discovery and Development

Binghe Wang, Series Editor

.....

Noora Sjöstedt,Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland

Tore B. Stage,Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark

.....

Добавление нового отзыва

Комментарий Поле, отмеченное звёздочкой  — обязательно к заполнению

Отзывы и комментарии читателей

Нет рецензий. Будьте первым, кто напишет рецензию на книгу Drug Transporters
Подняться наверх