265,99 €
A comprehensive explanation of drug metabolism concepts and applications in drug development and cancer treatment In the newly revised second edition of Drug Metabolism Handbook: Concepts and Applications in Cancer Research, a distinguished team of researchers delivers an incisive and robust exploration of the drug metabolism system and a well-illustrated and detailed explanation of the latest tools and techniques used in the research, pharmacology, and medicine. The book discusses the creation of new molecular entities, drug development, troubleshooting, and other highly relevant concepts, guiding readers through new applications in pharmaceutical research, development, and assessment. The latest edition offers updated content on metabolism basics and the application of a variety of new techniques to cancer treatment, including mass spectrometry, imaging, metabolomics, and immunotherapy. It also offers in-depth case studies highlighting the role of metabolism in drug development. Readers will also benefit from: * A thorough introduction to drug metabolism, including a historical perspective, factors affecting metabolism, and biotransformations in drug metabolism * Comprehensive discussions of technologies for in vitro and in vivo studies, including mass spectrometry and accelerating metabolite identification with mass spectrometry * In-depth explorations of drug interactions, including discussions of enzyme inhibition and the characterization of cytochrome P450 mechanism-based inhibition * Fulsome treatments of drug toxicity, including the role of drug metabolism in toxicity, and allergic reactions to drugs Perfect for medicinal chemists, pharmaceutical scientists, and toxicologists, Drug Metabolism Handbook: Concepts and Applications in Cancer Research, Second Edition will also earn a place in the libraries of analytical chemists and drug discovery professionals.
Sie lesen das E-Book in den Legimi-Apps auf:
Seitenzahl: 2100
Veröffentlichungsjahr: 2022
COVER
TITLE PAGE
COPYRIGHT
PREFACE
LIST OF CONTRIBUTORS
VOLUME 1
PART I: INTRODUCTION
CHAPTER 1: Historical Perspective
1.1 CONTROVERSIES SPANNING PAST, PRESENT, AND FUTURE
1.2 1800S: DISCOVERY OF MAJOR DRUG METABOLISM PATHWAYS (CONTI AND BICKEL, 1977)
1.3 1900–1950S: CONFIRMATION OF MAJOR PATHWAYS AND MECHANISTIC STUDIES
1.4 1950S–1980: MODERN DRUG METABOLISM EMERGES, WITH ENZYMATIC BASIS
1.5 1980–2005: FIELD DRIVEN BY IMPROVED TECHNOLOGIES
1.6 2005+: HIGH TECHNOLOGY
REFERENCES
CHAPTER 2: Factors Affecting Metabolism
REFERENCES
CHAPTER 3: Biotransformations in Drug Metabolism
3.1 DRUG METABOLISM IN DRUG DEVELOPMENT AND DRUG THERAPY
3.2 PREDICTION OF METABOLITE AND ENZYME RESPONSIBLE
3.3 FUNCTIONAL GROUP BIOTRANSFORMATIONS: PHASE I, PHASE II, AND CATALYSIS
3.4 OXIDATIONS AND CYTOCHROME P450
3.5 ENZYMOLOGY AND MODIFIERS OF CYTOCHROME P450S
REFERENCES
CHAPTER 4: A Comprehensive Picture of Biotransformation in Drug Discovery
4.1 INTRODUCTION
4.2 RATE OF METABOLISM
4.3 METABOLISM OF SMALL MOLECULES
4.4 ANALYTICAL TECHNOLOGIES IN DRUG METABOLISM
4.5 BIOTRANSFORMATION FOR NOVEL MODALITIES – PEPTIDES AND PROTEIN DEGRADERS
4.6 CONCLUSION
REFERENCES
CHAPTER 5:
In Vivo
Drug Metabolite Kinetics
5.1 INTRODUCTION
5.2
IN VIVO
DRUG METABOLITE KINETIC CONCEPTS AND PRINCIPLES
5.3 EFFECT OF INHIBITION AND INDUCTION ON METABOLITE KINETICS
5.4 DETERMINATION OF FORMATION AND ELIMINATION CLEARANCE OF METABOLITE
5.5 INCORPORATION OF PHARMACOLOGICALLY ACTIVE METABOLITE(S) IN PHARMACOKINETIC/PHARMACODYNAMIC MODELING
5.6 SUMMARY
ABBREVIATIONS
REFERENCES
CHAPTER 6: LC-MS/MS-Based Proteomics Methods for Quantifying Drug-Metabolizing Enzymes and Transporters
6.1 INTRODUCTION
6.2 MASS SPECTROMETRY VERSUS ALTERNATIVE PROTEIN QUANTIFICATION METHODS
6.3 MASS SPECTROMETRY DATA ACQUISITION METHODS FOR PROTEOMICS ANALYSIS
6.4 TARGETED APPROACHES
6.5 UNTARGETED PROTEOMICS APPROACHES
6.6 RELATIVE QUANTIFICATION VERSUS ABSOLUTE QUANTIFICATION
6.7 LABEL-BASED PROTEOMICS
6.8 LABEL-FREE PROTEOMICS
6.9 DMET PROTEIN QUANTIFICATION USING LC-MS/MS-BASED PROTEOMICS
6.10 POTENTIAL APPLICATION OF DMET EXPRESSION STUDIES
6.11 CONSIDERATIONS OF DMET PROTEIN QUANTIFICATION UTILIZING LC-MS/MS METHODS
6.12 CONCLUSION
REFERENCES
PART II: TECHNOLOGIES FOR
IN VITRO
AND
IN VIVO
STUDIES
CHAPTER 7: Mass Spectrometry
7.1 INTRODUCTION
7.2 A BRIEF HISTORY
7.3 THE MASS SPECTROMETRY LITERATURE
7.4 MASS SPECTROMETRY INSTRUMENTATION
7.5 INTERPRETATION: WHAT DOES IT MEAN
7.6 CONCLUSIONS
REFERENCES
NOTES
CHAPTER 8: Accelerating Metabolite Identification Mass Spectrometry Technology Drives Metabolite Identification Studies Forward
8.1 INTRODUCTION
8.2 CRITERIA FOR LC-MS METHODS
8.3 MATRICES EFFECT
8.4 TOOL OF CHOICE FOR METABOLITE CHARACTERIZATION
8.5 STRATEGIES FOR IDENTIFYING UNKNOWN METABOLITES
8.6 ONLINE HD-LC-MS
8.7 “ALL-IN-ONE” RADIOACTIVITY DETECTOR, STOP FLOW, AND DYNAMIC FLOW FOR METABOLITE IDENTIFICATION
8.8 METABOLIC ACTIVATION STUDIES BY MASS SPECTROMETRY
8.9 STRATEGIES TO SCREEN FOR REACTIVE METABOLITES
8.10 SUMMARY
ABBREVIATIONS AND GLOSSARY
REFERENCES
CHAPTER 9: Role of Structural Modifications of Drug Candidates to Enhance Metabolic Stability
9.1 BACKGROUND
9.2 INTRODUCTION
9.3 SIGNIFICANCE OF METABOLITE CHARACTERIZATION AND STRUCTURE MODIFICATION
9.4 ENHANCE METABOLIC STABILITY
9.5 METABOLIC STABILITY AND INTRINSIC METABOLIC CLEARANCE
9.6 ADVANTAGES OF ENHANCING METABOLIC STABILITY
9.7 STRATEGIES TO ENHANCE METABOLIC STABILITY
9.8 ANALYTICAL TOOLS
9.9 CASE STUDIES
9.10 CONCLUSIONS
REFERENCES
CHAPTER 10: Drug Design Strategies: Role of Structural Modifications of Drug Candidates to Improve PK Parameters of New Drugs
10.1 ACTIVE METABOLITES
10.2 ORAL ABSORPTION AND INTRAVENOUS DOSE
10.3 PK ANALYSIS
10.4 CASE STUDIES
10.5 PRODRUGS TO INCREASE WATER SOLUBILITY
10.6 CONCLUSION
REFERENCES
CHAPTER 11: Chemical Structural Alert and Reactive Metabolite Concept as Applied in Medicinal Chemistry to Minimize the Toxicity of Drug Candidates
11.1 IMPORTANCE OF REACTIVE INTERMEDIATES IN DRUG DISCOVERY AND DEVELOPMENT
11.2 IDIOSYNCRATIC DRUG TOXICITY AND MOLECULAR MECHANISMS
11.3 KEY TOOLS AND STRATEGIES TO IMPROVE DRUG SAFETY
11.4 PEROXIDASES
11.5 ACYL GLUCURONIDATION AND
S
-Acyl-CoA Thioesters
11.6 COVALENT BINDING
11.7 MECHANISTIC STUDIES
11.8 PRECLINICAL DEVELOPMENT
11.9 CLINICAL DEVELOPMENT: STRATEGY
11.10 CASE STUDIES
11.11 CONCLUSION AND FUTURE POSSIBILITIES
REFERENCES
CHAPTER 12: Studies of Reactive Metabolites using Genotoxicity Arrays and Enzyme/DNA Biocolloids – 2021
12.1 INTRODUCTION
12.2 ON DEMAND METABOLIC REACTIONS
12.3 ARRAYS WITH ELECTROCHEMICAL DETECTION
12.4 ELECTROCHEMILUMINESCENT ARRAYS
12.5 ECL ARRAYS CAN MEASURE BOTH DNA OXIDATION AND NUCLEOBASE ADDUCTION
12.6 DETECTING SITE-SPECIFIC DAMAGE TO
TUMOR SUPPRESSOR
GENES
12.7 EMERGING TECHNOLOGIES AND METHODS
12.8 CONCLUSIONS AND FUTURE OUTLOOK
ACKNOWLEDGMENTS
BIOGRAPHIES
REFERENCES
PART III: DRUG INTERACTIONS
CHAPTER 13: Enzyme Inhibition
13.1 INTRODUCTION
13.2 MECHANISMS OF ENZYME INHIBITION
13.3 COMPETITIVE INHIBITION
13.4 NONCOMPETITIVE INHIBITION
13.5 UNCOMPETITIVE INHIBITION
13.6 PRODUCT INHIBITION
13.7 TRANSITION-STATE ANALOGS
13.8 SLOW, TIGHT-BINDING INHIBITORS
13.9 MECHANISM-BASED INACTIVATORS
13.10 INHIBITORS THAT ARE METABOLIZED TO REACTIVE PRODUCTS THAT COVALENTLY ATTACH TO THE ENZYME
13.11 SUBSTRATE INHIBITION
13.12 PARTIAL INHIBITION
13.13 INHIBITION OF CYTOCHROME P450 ENZYMES
13.14 REVERSIBLE INHIBITORS
13.15 QUASI-IRREVERSIBLE INHIBITORS
13.16 MECHANISM-BASED INACTIVATORS
REFERENCES
CHAPTER 14: Xenobiotic Receptor-Mediated Gene Regulation in Drug Metabolism and Disposition
14.1 INTRODUCTION
14.2 PREGNANE X RECEPTOR
14.3 CONSTITUTIVE ANDROSTANE/ACTIVATED RECEPTOR (CAR)
14.4 CLOSING REMARKS AND PERSPECTIVES
ACKNOWLEDGMENTS
REFERENCES
CHAPTER 15: Characterization of Cytochrome P450 Mechanism Based Inhibition
15.1 INTRODUCTION
15.2 INHIBITORS THAT UPON ACTIVATION BIND COVALENTLY TO THE P450 APOPROTEIN
15.3 INHIBITORS THAT INTERACT IN A PSEUDOIRREVERSIBLE MANNER WITH HEME IRON
15.4 INACTIVATION THAT CAUSE DESTRUCTION OF THE PROSTHETIC HEME GROUP, OFTEN TIMES LEADING TO HEME-DERIVED PRODUCTS THAT COVALENTLY MODIFY THE APOPROTEIN
REFERENCES
CHAPTER 16: An Introduction to Metabolic Reaction-Phenotyping
16.1 INTRODUCTION
16.2 SIGNIFICANT DRUG-METABOLIZING ENZYMES
16.3 COMMON
IN VITRO
METHODS TO ASSESS DRUG METABOLISM
16.4
IN VITRO
TO
IN VIVO
EXTRAPOLATION OF METABOLIC CLEARANCE
16.5 SUMMARY
REFERENCES
CHAPTER 17: Epigenetic Regulation of Drug-Metabolizing Enzymes in Cancer
17.1 INTRODUCTION
17.2 DNA METHYLATION OF DME
17.3 HISTONE MODIFICATION
17.4 NONCODING RNA
17.5 RNA METHYLATION
17.6 CLOSING REMARKS AND PERSPECTIVES
ACKNOWLEDGMENTS
REFERENCES
CHAPTER 18: Epigenetic Regulation of Drug Transporters in Cancer
18.1 INTRODUCTION
18.2 DNA METHYLATION
18.3 HISTONE MODIFICATIONS
18.4 NONCODING RNA
18.5 Closing Remarks and Perspectives
ACKNOWLEDGMENTS
REFERENCES
VOLUME 2
PART IV: TOXICITY
CHAPTER 19: The Role of Drug Metabolism in Toxicity
19.1 INTRODUCTION
19.2 DRUG METABOLIZING ENZYMES
19.3 CLASSIFICATION OF TOXICITY
19.4 MOLECULAR MECHANISMS OF TOXICITY
19.5 ORGAN SYSTEMS TOXICOLOGY
19.6 CARCINOGENESIS
19.7 TERATOGENESIS
19.8 ABROGATION/MITIGATION OF BIOACTIVATION – CASE EXAMPLES
19.9 EXPERIMENTAL METHODS FOR SCREENING
19.10 SUMMARY
ACKNOWLEDGMENT
References
CHAPTER 20: Allergic Reactions to Drugs
20.1 INTRODUCTION
20.2 IMMUNE SYSTEM: A BRIEF OVERVIEW
20.3 DRUG METABOLISM AND THE HAPTEN HYPOTHESIS
20.4 ALLERGIC REACTIONS TO DRUGS (EXAMPLES)
20.5 CONCLUSION
ACKNOWLEDGMENTS
REFERENCES
NOTE
CHAPTER 21: Chemical Mechanisms in Toxicology
21.1 INTRODUCTION
21.2 GLUTATHIONE ADDUCTS
21.3 COVALENT BINDING
21.4 STRUCTURAL ALERTS
21.5 EXAMPLES OF METABOLIC ACTIVATION
21.6 CONCLUSION
ACKNOWLEDGMENTS
REFERENCES
NOTE
CHAPTER 22: Role of Bioactivation Reactions in Chemically Induced Nephrotoxicity
22.1 TOXICOLOGICAL IMPLICATIONS OF RENAL STRUCTURE AND FUNCTION
22.2 INTERORGAN AND INTRARENAL BIOACTIVATION PATHWAYS
22.3 CYTOCHROME P450 (CYP)-DEPENDENT BIOACTIVATION IN THE RENAL PROXIMAL TUBULE
22.4 FLAVIN-CONTAINING MONOOXYGENASE (FMO)-DEPENDENT BIOACTIVATION IN THE RENAL PROXIMAL TUBULE
22.5 GLUTATHIONE (GSH)-DEPENDENT BIOACTIVATION IN THE RENAL PROXIMAL TUBULE
22.6 RENAL METABOLISM OF ACETAMINOPHEN (PARACETAMOL) BY THE KIDNEYS
22.7 IMPACT OF CHRONIC KIDNEY DISEASE ON DRUG METABOLISM
22.8 SUMMARY AND CONCLUSION
REFERENCES
PART V: APPLICATIONS
CHAPTER 23: Mapping the Heterogeneous Distribution of Cancer Drugs by Imaging Mass Spectrometry
23.1 INTRODUCTION
23.2 MSI INSTRUMENTATION
23.3 IMAGE ANALYSIS
23.4 SAMPLE PREPARATION
23.5 QUANTITATIVE MSI
23.6 MSI IN STUDYING THE DISTRIBUTION OF CANCER DRUGS
23.7 EMERGING IN VITRO APPLICATIONS
23.8 CLINICAL TRANSLATION
23.9 CONCLUSION AND PERSPECTIVES
REFERENCES
CHAPTER 24: Systemic Metabolomic Changes Associated with Chemotherapy: Role in Personalized Therapy
24.1 INTRODUCTION
24.2 METABOLOMICS: MAPPING METABOLOMIC ALTERATIONS IN CANCER
24.3 ANALYTICAL METHODS IN CANCER METABOLOMICS: GLOBAL AND TARGETED STRATEGIES
24.4 METABOLOMICS IN CANCER: EXPLORATION OF METABOLIC ALTERATIONS DURING DISEASE DEVELOPMENT, CHEMOTHERAPY AND DRUG RESISTANCE
24.5 CHALLENGES AND PROGRESS IN THE APPLICATION OF METABOLOMICS FOR THE DEVELOPMENT OF PERSONALIZED CANCER MEDICINE
24.6 FUTURE PERSPECTIVES AND FURTHER SCOPE OF IMPROVEMENT IN METABOLOMICS-ASSISTED CANCER THERAPY
24.7 CONCLUDING REMARKS
REFERENCES
CHAPTER 25: Metabolic Reprogramming in Cancer
25.1 INTRODUCTION
25.2 REPROGRAMMING OF CENTRAL CARBON METABOLISM (CCM)
25.3 GLUTAMINE ADDICTION OF CANCER CELLS
25.4 ARGININE AND POLYAMINE METABOLISM
25.5 SERINE, METHIONINE AND ONE CARBON METABOLISM
25.6 BRANCHED-CHAIN AMINO ACID (BCAA) METABOLISM
25.7 REPROGRAMMING OF OXIDATIVE STRESS RESPONSE MACHINERY
25.8 BIOSYNTHESIS OF PURINE AND PYRIMIDINE NUCLEOTIDES IN CANCER
25.9 REPROGRAMMING OF LIPID METABOLISM IN CANCER
25.10 METABOLIC REWIRING IN CANCER STEM CELLS (CSC)
25.11 FUTURE PERSPECTIVES
REFERENCES
CHAPTER 26: Case Study: Metabolism and Reactions of Alkylating Agents in Cancer Therapy
26.1 INTRODUCTION
26.2
IN VITRO
STUDIES
26.3 NOVEL REARRANGEMENT
26.4 IDENTIFICATION AND CHARACTERIZATION OF
IN VITRO
METABOLITE/DECOMPOSITION PRODUCTS OF LAROMUSTINE IN HLM
26.5 IDENTIFICATION AND CHARACTERIZATION OF
IN VITRO
CONJUGATION REACTIONS OF LAROMUSTINE
26.6 DISCUSSION
REFERENCES
CHAPTER 27: Rewiring of Drug Metabolism and Its Cross-talk with Metabolic Reprogramming in Cancer
27.1 INTRODUCTION
27.2 PATHWAYS INVOLVED IN ANTICANCER DRUG METABOLISM
27.3 ALTERATIONS IN DRUG METABOLISM IN CANCER
27.4 REWIRING OF DRUG METABOLISM IN RESPONSE TO ANTICANCER AGENTS
27.5 CROSS-TALK BETWEEN ENDOGENOUS AND DRUG METABOLISM IN CANCER
27.6 ROLE OF MICROENVIRONMENT IN DRUG METABOLISM
27.7 FUTURE PERSPECTIVES
ACKNOWLEDGMENT
REFERENCES
CHAPTER 28: Principles of Drug Metabolism and Interactions in Cardio-Oncology
28.1 INTRODUCTION
28.2 EXPRESSION OF CYP450 IN CANCER
28.3 EXPRESSION OF P-GLYCOPROTEIN IN CANCER
28.4 INTERACTIONS BETWEEN DRUG CLASSES IN CARDIO-ONCOLOGY BY CYP450 OR P-G
28.5 IMPACT OF GENOMIC VARIATION AND OTHER FORMS OF REGULATION
28.6 MULTIDISCIPLINARY TEAM APPROACH
28.7 CONCLUSION
REFERENCES
INDEX
END USER LICENSE AGREEMENT
Chapter 1
TABLE 1.1 Early development of the major drug metabolism pathways.
Chapter 3
TABLE 3.1 Effective uses of metabolism science in drug development and drug ...
TABLE 3.2 Major P450 reactions classified according to category and mechanis...
TABLE 3.3 Regioselectivity preferences of CYP3A4, CYP2D6, and CYP2C9.
Chapter 4
TABLE 4.1 Scaling factors using to extrapolate in vitro clearance to in vivo...
TABLE 4.2 Use of deuterium atoms to reduce the C—H bond cleavage via Kinetic...
TABLE 4.3 Comparison of P450 and aldehyde oxidase enzyme systems.
TABLE 4.4 List of important UGTs present in humans and their substrates.
TABLE 4.5 Comparison of mass analyzer with respect to key performance attrib...
TABLE 4.6 Common derivatization techniques for various functional groups.
TABLE 4.7 Mass shifts and mass defects of conjugated metabolites.
Chapter 5
TABLE 5.1 Characteristics and human half-life of some approved antibody-drug...
Chapter 7
TABLE 7.1 Example calculations of relative molecular mass.
TABLE 7.2 Accurate masses and natural abundances of selected stable isotopes...
TABLE 7.3 Elemental compositions consistent with an experimental accurate ma...
TABLE 7.4 Calculation of average molecular mass and polydispersity for Trito...
Chapter 8
TABLE 8.1 Common biotransformation reactions.
TABLE 8.2 Techniques that help in the search for, and identification of, met...
TABLE 8.3 Exchange of labile hydrogens in nimodipine and metabolites formed ...
Chapter 9
TABLE 9.1 Enhancement of metabolic stability through structural modification...
TABLE 9.2 Tools for the identification and characterization of metabolites....
Chapter 10
TABLE 10.1 Metabolism-driven optimization of PK properties by structure modi...
Chapter 11
TABLE 11.1 Examples of chemical structures activating to produce toxic metab...
TABLE 11.2 Selected tools to screen for reactive metabolites, covalent bindi...
Chapter 15
TABLE 15.1 GSH and other nucleophiles used in trapping electrophilic interme...
TABLE 15.2 Average enzyme abundances taken from SimP450 V7.1, illustrating t...
TABLE 15.3 A table of in vitro IC50 generated against three different P450 3...
Chapter 16
TABLE 16.1 Survey of drug-metabolizing enzymes mediating oxidation or reduct...
TABLE 16.2 Human cytochromes P450 and their substrate selectivity.
TABLE 16.3 Tissue distribution and substrates of major human uridine 5′-diph...
TABLE 16.4 Cytochrome P450 (CYP) substrates, inhibitors, and inducers (Unite...
TABLE 16.5 Common chemical inhibitors and recommended concentrations for in ...
TABLE 16.6 Examples of known substrates for the major human flavin-containin...
TABLE 16.7 Meta-analysis of hepatic cytochrome P450 (CYP) abundance.
TABLE 16.8 Examples of an integrated in vitro cytochrome P450 (CYP) reaction...
TABLE 16.9 Examples of in vitro cytochrome P450 (CYP) reaction phenotyping o...
Chapter 19
TABLE 19.1 Families and major distribution of common human drug-metabolizing...
TABLE 19.2 Functional groups and drugs associated with reactive metabolite f...
TABLE 19.3 Drugs, putative reactive-intermediates, implicated enzymes, and a...
TABLE 19.4 Structural modification and mitigation of toxicity.
Chapter 20
TABLE 20.1 Partial list of carboxylic-acid containing drugs withdrawn from c...
Chapter 21
TABLE 21.1 Hard
versus
soft nucleophiles and electrophiles.
TABLE 21.2 Drugs or chemicals known to cause idiosyncratic hepatotoxicity....
TABLE 21.3 Examples of chemicals containing structural alerts and their corr...
Chapter 22
TABLE 22.1 Major cytochrome P450 (CYP) enzymes in rat and human kidney.
TABLE 22.2 Mammalian enzymes that catalyze cysteine conjugate β-lyase activi...
TABLE 22.3 Summary of some parameters altered in chronic kidney disease (CKD...
Chapter 23
TABLE 23.1 Use of MSI for studying the distribution of cancer drugs.
Chapter 24
TABLE 24.1 Metabolic alterations associated with chemotherapeutic drug resis...
Chapter 26
TABLE 26.1 Formation of C-7 and loss of substrate in incubations of [14C]lar...
TABLE 26.2 Results from Fourier transform ion cyclotron resonance-mass spect...
TABLE 26.3 Metabolite/decomposition products formed in HLM incubation.
TABLE 26.4
In vitro
conjugation reactions of laromustine and VNP4090CE.
Chapter 27
TABLE 27.1 Chemotherapy drugs and their modes of actions.
TABLE 27.2 Cytochrome P450s involved in cancer drug metabolism.
TABLE 27.3 Altered expression of Cytochrome P450s in cancer and its impact....
TABLE 27.4 UGTs and GSTs involved in cancer drug metabolism.
TABLE 27.5 Altered expression of UGTs in cancer and its impact on drug respo...
TABLE 27.6 Association between alteration in GST expression and therapeutic ...
Chapter 28
TABLE 28.1 Various cancer and anticoagulation drug classes associate with dru...
Chapter 1
Figure 1.1 Examples of first-observed metabolism reactions. (a) glycine conj...
Chapter 3
Figure 3.1 Role of drug metabolism in bioavailability and clearance.
Figure 3.2 Example of effective use of metabolism science: fexofenadine (All...
Figure 3.3 Examples of phase I reactions.
Figure 3.4 Examples of phase II reactions.
Figure 3.5 Relationships and nomenclature of cytochrome P450 members importa...
Figure 3.6 Average relative proportions of CYPs present in human liver.
Figure 3.7 Carbon hydroxylation under the unified mechanism. (i) H
⋅
ab...
Figure 3.8 Order of reactivity of sp
3
carbons toward cytochrome P450 oxidati...
Figure 3.9 Examples of carbon hydroxylation. (a) Benzylic carbon hydroxylati...
Figure 3.10 Mechanism for oxidation of ethanol and hydrated acetaldehyde by ...
Figure 3.11 Oxidation of sp
2
systems to form arene oxide or epoxide.
Figure 3.12 Examples of olefin (alkene) epoxidation and aromatic hydroxylati...
Figure 3.13 Unified P450 mechanism for N-dealkylation of amine nitrogen.
Figure 3.14 Unified P450 mechanism for O-dealkylation of ethers (a) and oxid...
Figure 3.15 Examples of N-dealkylation, O-dealkylation, and oxidative cleava...
Figure 3.16 Unified P450 mechanism for N-oxygenation.
Figure 3.17 Examples of nitrogen oxidation. (a) Primary amine oxidation, (b)...
Figure 3.18 Examples of sulfur oxidation.
Figure 3.19 Other oxidations relevant to drug metabolism.
Figure 3.20 Decision tree for prediction of dominant cytochrome P450 isoform...
Figure 3.21 Binding cavities of human cytochrome P450 members (show my own d...
Chapter 4
Figure 4.1 Representation of various liver systems that can be utilized to s...
Figure 4.2 List of typical reactions mediated by P450s.
Figure 4.3 Catalytic cycle for Cytochrome P450.
Figure 4.4 Relative amounts of P450s present in humans.
Figure 4.5 Effect of intrinsic clearance by reducing lipophilicity.
Figure 4.6 Example for blocking the site of metabolism by incorporation of f...
Figure 4.7 Example scaffold hopping to reduce metabolic liability.
Figure 4.8 Partial list of chemical scaffolds that can be metabolized by ald...
Figure 4.9 Catalytic cycle of aldehyde oxidase.
Figure 4.10 Catalytic cycle for flavin-dependent monooxygenase (FMOs).
Figure 4.11 The steps involved in the reduction of a nitro group to an amino...
Figure 4.12 (a) UPLC/TopCount radiochromatogram of the standard mixture (1, ...
Figure 4.13 Flowchart of optimized acquisition sequence method with backgrou...
Figure 4.14 In-depth structure elucidation of a low abundant ticlopidine GSH...
Figure 4.15 Proposed structures of pioglitazone metabolites in dog kidney an...
Figure 4.16 LC/MS analysis of pyridine and piperidine N-oxide in urine befor...
Figure 4.17 Aminolysis reaction showing nucleophilic displacement of glucuro...
Figure 4.18 Common mass defect filter templates and various classes of metab...
Figure 4.19 TIC and radiochromatogram of rat plasma following a single oral ...
Figure 4.20 (a) Unprocessed and (b) isotope filtering processed mass spectra...
Figure 4.21 Shown is the progression of GLP-1 receptor agonists and the chem...
Figure 4.22 Shown are all states of a solution NMR structure of Hepcidin (PD...
Figure 4.23 Synthetically and biosynthetically made crosslinks observed in (...
Figure 4.24 Both 2-pyridine carboxaldehyde derivatives (a) and 2-methylthioa...
Figure 4.25 Structures of commonly used IMIDs are shown (a) with a heat map ...
Chapter 5
Figure 5.1 A schematic representation of the fate of a metabolite (X) in the...
Figure 5.2 A metabolite kinetic model.
Figure 5.3 A hypothetical example that illustrates the conceptual difference...
Figure 5.4 Effects of the rate-limiting step in metabolite kinetics on the s...
Figure 5.5 Effects of the rate-limiting step in metabolite kinetics on the t...
Figure 5.6 The serum concentration–time profiles of trastuzumab deruxtecan (...
Figure 5.7 Effects of presystemically formed metabolites on the shape of met...
Figure 5.8 The plasma concentration–time profiles of midazolam and 1′-hydrox...
Figure 5.9 A representative hybrid physiologically based pharmacokinetic mod...
Figure 5.10 A kinetic model for sequential metabolism. CL
e
, CL
e
(m1), and CL
e
Figure 5.11 Inhibition of the formation clearance of a metabolite on metabol...
Figure 5.12 Sensitivity of various pharmacokinetic parameters to the inhibit...
Chapter 6
Figure 6.1 Schematic presentation of the data acquisition methods SRM/MRM, P...
Figure 6.2 Comparative bar graph contrasting the different data acquisition ...
Figure 6.3 Hepatic drug-metabolizing enzymes (a) and transporters (b) (DMET)...
Figure 6.4 Intestinal drug metabolizing enzymes (a) and transporters (b) (DM...
Figure 6.5 Renal drug metabolizing enzymes (a) and transporters (b) (DMET) e...
Chapter 7
Figure 7.1 Modular diagram of a mass spectrometer system. Upper panel is ill...
Figure 7.2 Applicability of various chromatography-mass spectrometry techniq...
Figure 7.3 Combining liquid chromatography with mass spectrometry involves a...
Figure 7.4 Schematic diagram of a moving belt LC-MS interface.
Figure 7.5 Electrospray ion source.
Figure 7.6 Comparison of EI mass spectra of hexadecane. The 1951 mass spectr...
Figure 7.7 Electrospray ionization mechanism.
Figure 7.8 Applicable range of molecules ionizable by APPI.
Figure 7.9 Desorption electrospray ionization source.
Figure 7.10 MS-MS operation modes.
Figure 7.11 Chromatography-mass spectroscopy data space.
Figure 7.12 Selected reaction monitoring MS-MS operation mode.
Figure 7.13 LC-MS separation of PFAAs, PFASs, fluorotelomers and others. Dia...
Figure 7.14 Fragmentations of classes of perfluoroalkanes. Top: perfluoroalk...
Figure 7.15 ESI mass spectrum of a molecule containing two chlorine atoms.
Figure 7.16 Comparison of the isotope pattern for the observed electrospray ...
Figure 7.17 Boron isotope patterns.
Figure 7.18 Observed isotope pattern (upper panel) and predicted model (lowe...
Structure 1
Figure 7.19 MS-MS product ion spectra of the
m/z
300 (top panel),
m/z
302 (m...
Figure 7.20 Partial accurate mass spectrum, comparing mass resolution calcul...
Figure 7.21 Example of high-resolution experiment on complex mixtures, showi...
Figure 7.22 Example of a high resolution experiment on complex mixtures, sho...
Figure 7.23 MS-MS product ion spectra of the
m/z
300 [M+H] at 10V (top ...
Figure 7.24 Top panel: ESI mass spectrum of sertraline. Bottom panel: ESI MS...
Scheme 7.1 ESI fragmentation of sertraline.
Structure 2
Figure 7.25 Positive ion ESI mass spectrum of a phosphatidylcholine, taken f...
Figure 7.26 Differentiation of keto (left structure and mass spectrum) and e...
Structure 3
Figure 7.27 Hydrogen–deuterium exchange of melamine in a DART ion source....
Structure 4
Figure 7.28 Positive ion electrospray mass spectrum of the molecular ion reg...
Figure 7.29 Deuterium exchanged molecular ion region of anidulafungin. (From...
Figure 7.30 Electrospray spectrum of sodium iodide clusters. The spectrum is...
Figure 7.31 Formation of different adduct ions under different solvent condi...
Structure 5
Figure 7.32 Positive ion ESI mass spectrum of a poly(ethylene glycol) mixtur...
Figure 7.33 Fast atom bombardment spectrum of a cetirizine poly(ethylene gly...
Structure 6
Structure 7
Figure 7.34 Electrospray mass spectrum of octoxynol (triton X).
Figure 7.35 Comparison of the isotope pattern of the doubly charged molecula...
Figure 7.36 Comparison of the isotope pattern of the singly charged molecula...
Figure 7.37 Positive ion ESI mass spectrum of bovine serum albumin. The aver...
Figure 7.38 Amino acid sequence of insulin.
Figure 7.39 Electrospray mass spectrum of insulin. Top panel: the multiply c...
Scheme 7.2 Reversible Schiff base formation between
o
-vanillin and the ε-ami...
Figure 7.40 Multiply charged ESI mass spectrum of porcine somatotropin (PST)...
Figure 7.41 Multiply charged electrospray mass spectrum of OV-PST.
Figure 7.42 MaxEnt®-transformed singly charged mass spectrum of OV-PST. This...
Chapter 8
Figure 8.1 The chemical structure of nimodipine with the labile H exchange a...
Figure 8.2 QTOF MS/MS spectra of metabolite M-3 in (a) D
2
O and (b) H
2
O.
Figure 8.3 Proposed metabolic pathways of nimodipine in hepatic microsomal i...
Figure 8.4 Proposed metabolic pathways of nimodipine in hepatic microsomal i...
Figure 8.5 Hardware schematic diagram of the LC-ARC system. Source: AIM Rese...
Figure 8.6 Chemical structure of [14C]dextromethorphan, the test compound us...
Figure 8.7 HPLC-MS chromatograms of [14C]dextromethorphan following incubati...
Figure 8.8 Representative LC-MS spectra of [M+H]+
m
/
z
274, M-1, (a) MS
2
, (b)...
Figure 8.9 Proposed metabolic pathways of dextromethorphan in human hepatic ...
Chapter 11
Figure 11.1 Reactive intermediate theory and idiosyncratic reactions/toxic e...
Chapter 12
Figure 12.1 Conceptual representation of films of DNA, polyions, and metabol...
Scheme 12.1 Polyions used to make films by layer-by-layer (LbL) alternate el...
Scheme 12.2 Microfluidic array system used for electrochemical detection of ...
Figure 12.2 Representative SWVs for oxidation of damaged DNA at different en...
Scheme 12.3 DNA adduct formation from metabolic cyt P450 bioactivation of (a...
Figure 12.3 Influence of substrate incubation time on SWV peak current ratio...
Scheme 12.4 Microfluidic ECL chip for multiple-enzyme screening of metabolit...
Scheme 12.5 Experimental design of 64 nanowell ECL array with films of enzym...
Scheme 12.6 Proposed metabolic pathways of NNK in humans.
Figure 12.4 ECL data from array wells containing Ru
II
PVP/enzyme/DNA films re...
Figure 12.5 Single reaction monitoring (SRM) LC-MS chromatograms for
m
/
z
299...
Figure 12.6 Automated genotoxicity screening array: (a) The 3D-printed devic...
Figure 12.7 ECL array results comparing extracted vapor from e-cigarettes wi...
Scheme 12.7 Strategy for detecting DNA damage using (a) DNA/enzyme films, (b...
Scheme 12.8 Major metabolic pathway of arylamines causing DNA adduct formati...
Scheme 12.9 Proposed pathway involving Cu
2+
and NADPH for ROS formation ...
Figure 12.8 Recolorized and reconstructed images of ECL for DNA oxidation (a...
Figure 12.9 Bar graphs showing (a) relative DNA oxidation rate and (b) relat...
Figure 12.10 An overview of the sequence-specific DNA hybridization damage e...
Chapter 14
Figure 14.1 Structure and functional domains of nuclear receptors. (a) Basal...
Figure 14.2 Schematic representation of the creation of hPXR “humanized” mic...
Figure 14.3 Newly proposed model of cross-talk between PXR and CAR in the re...
Chapter 15
Figure 15.1 A graphical breakdown of the various pathways that contribute to...
Figure 15.2 A simple graph depicting an imaginary time concentration profile...
Figure 15.3 A simple schematic describing the possible mechanisms of inhibit...
Figure 15.4 A schematic diagram showing the free-energy profile of the cours...
Figure 15.5 The catalytic cycle for cytochrome P450 enzyme oxidation reactio...
Figure 15.6 A brief listing of common P450 oxidation reactions.
Figure 15.7 Reaction scheme of a mechanism-based enzyme inactivator.
Figure 15.8 Multiple P450 3A4 oxidation reactions toward a single substrate,...
Figure 15.9 Mechanistic scheme for P450 enzyme-mediated bioactivation and su...
Figure 15.10 Mechanistic scheme for P450 enzyme-mediated bioactivation and s...
Figure 15.11 Mechanistic scheme for P450 enzyme-mediated bioactivation and s...
Figure 15.12 Mechanistic scheme for P450 enzyme-mediated bioactivation and s...
Figure 15.13 Mechanistic scheme for P450 enzyme-mediated bioactivation and s...
Figure 15.14 Mechanistic scheme for P450 enzyme-mediated bioactivation and s...
Figure 15.15 SimP450-generated single oral dose pharmacokinetic model of mid...
Figure 15.16 Represents a schematic of the MBI testing funnel describing the...
Figure 15.17 The absolute energetic differences between different P450-catal...
Figure 15.18 Crystal structure of P450 2C9 with flurbiprofen bound (1R90.pdb...
Figure 15.19 Hypothetical drug molecule metabolized by two different P450 en...
Figure 15.20 P450 active sites have unique active site which may contribute ...
Figure 15.21 The structure of psoralen, 8-methoxypsoralen, and 5-methoxypsor...
Figure 15.22 Mechanistic scheme for P450 enzyme-mediated bioactivation of tr...
Figure 15.23 Mechanistic scheme for P450 enzyme-mediated bioactivation of ar...
Figure 15.24 Data transformed from the experimental design in Figure 15.25 c...
Figure 15.25 The 96-well plate preincubation format for determination of IC
5
...
Figure 15.26 Different plots for time-dependent inhibition data.
Figure 15.27 A 96-well plate preincubation format for determination of time-...
Figure 15.28 Kinetic formation of MIC by monitoring absorbance at 455 nm....
Figure 15.29 Measurement of carbon monoxide binding pre- and postincubation ...
Figure 15.30 Mechanistic scheme for P450 enzyme-mediated bioactivation and s...
Figure 15.31 LC-MS spectra and deconvoluted spectra for P450 3A4 apoprotein....
Figure 15.32 (a) Chromatogram illustrating separation of reductase and apopr...
Figure 15.33 Different mechanistic approaches to locating P450 nucleophiles ...
Figure 15.34 Chromatographic separation of peptides following proteinase K d...
Figure 15.35 (a) Covalent binding of EE to P450 3A5. SDS-PAGE separation of ...
Figure 15.36 A structural comparison of two compounds aimed as selective est...
Figure 15.37 Kinetic equations used to extrapolate
in vitro
MBI into clinica...
Chapter 19
Figure 19.1
Reactivity scheme for acyl glucuronides
. Acyl glucuronide can re...
Figure 19.2 Reactivity scheme for
N
-acetyl transferases.
Figure 19.3 Glutathione redox cycle.
Figure 19.4
General scheme of toxicity
. Toxicity of a drug and its metabolit...
Chapter 20
Figure 20.1 Scheme for the metabolism of drugs to chemically-reactive interm...
Figure 20.2 Chemical mechanisms for the covalent binding of penicillin to pr...
Figure 20.3 Proposed chemical mechanism for the metabolic activation of felb...
Figure 20.4 Chemical mechanism for the P450-mediated bioactivation of haloth...
Figure 20.5 Proposed chemical mechanism for the metabolic activation of tien...
Figure 20.6 Chemical mechanism for the P450-mediated bioactivation of sulfam...
Figure 20.7 Proposed scheme for the metabolic activation of carboxylic acid-...
Figure 20.8 Mechanism for transacylation of protein nucleophiles by 1-β-
O
-ac...
Figure 20.9 Mechanism for stable adduct formation with lysine-residues on pr...
Figure 20.10 Proposed chemical mechanism for the metabolic activation of ben...
Figure 20.11 Risk of allergic reactions to drugs (circles) and corresponding...
Chapter 21
Figure 21.1 Metabolic activation of benzo[
a
]pyrene leading to covalent bindi...
Figure 21.2 Metabolic activation of xenobiotics to chemically-reactive and p...
Figure 21.3 Chemically-reactive compounds react with GSH by one of two gener...
Figure 21.4 Tandem mass spectrometric LC-MS/MS screening for glutathione add...
Figure 21.5 Enzyme-mediated degradation of glutathione conjugates in the kid...
Figure 21.6 Tandem mass spectrometric LC-MS/MS screening for mercapturic aci...
Figure 21.7 Scheme for the metabolic activation of naphthalene and formation...
Figure 21.8 Scheme for the metabolic activation of suprofen to electrophilic...
Figure 21.9 Scheme for the metabolic activation of furosemide to reactive fu...
Figure 21.10 Scheme for the metabolic activation of the thiazolidinedione mo...
Figure 21.11 Scheme for the metabolic activation of bromobenzene and formati...
Figure 21.12 Proposed scheme for the metabolic activation of the raloxifene ...
Figure 21.13 Scheme for the metabolic activation of zafirlukast to an electr...
Figure 21.14 Proposed scheme for the concerted metabolic activation of valpr...
Figure 21.15 Proposed scheme for the metabolic activation of the zomepirac b...
Figure 21.16 Proposed scheme for the metabolic activation of the zomepirac a...
Figure 21.17 Proposed scheme for the metabolic activation of procarbazine le...
Figure 21.18 Proposed mechanism in the metabolic activation of isoniazid....
Figure 21.19 Proposed mechanism in the metabolic activation of acetaminophen...
Figure 21.20 Proposed mechanism of Nrf2 activation by reactive oxygen specie...
Figure 21.21 Proposed mechanisms in the metabolic activation of diclofenac....
Figure 21.22 Proposed mechanisms in the metabolic activation of the ovotoxin...
Figure 21.23 Proposed mechanisms in the metabolic activation of the estrogen...
Figure 21.24 Proposed mechanism in the metabolic activation of dapsone to a ...
Figure 21.25 Proposed mechanism in the metabolic activation of nitrobenzene ...
Chapter 22
Figure 22.1 Scheme of interorgan pathways involving cytochrome P450 (CYP) en...
Figure 22.2 Scheme of interorgan pathways involving GSH conjugation. Xenobio...
Figure 22.3 Bioactivation of GSH conjugates. Drug or xenobiotic (R-X) forms ...
Figure 22.4 Structures of representative cysteine conjugates that undergo re...
Figure 22.5 Validation of role of enzymatic and transport processes in the r...
Figure 22.6 Renal metabolism of acetaminophen (APAP). APAP may undergo eithe...
Chapter 23
Figure 23.1 Schematic presentation of (a) matrix-assisted laser desorption/i...
Figure 23.2 Workflow in recording the ion images of a tissue section. The io...
Figure 23.3 A typical MSI workflow in pharmaceutical research shows the step...
Figure 23.4 The top panel shows the MALDI-MSI data revealing the distributio...
Figure 23.5 Histological localization of the drug (epacadostat) and metaboli...
Figure 23.6 Temporal profile of enzalutamide (ENZ) penetration in an
ex vivo
Figure 23.7 MALDI-MSI showing the temporal profile of irinotecan (
m/z
587) p...
Figure 23.8 Detection of cetuximab, a monoclonal antibody, using MALDI-MSI i...
Figure 23.9 (a) Ion images of cetuximab distribution in treated and control ...
Chapter 24
Figure 24.1 A schematic diagram of the personalized metabolomics workflow f...
Chapter 25
Figure 25.1 Schematic representation of glucose metabolism in (a) normal ver...
Figure 25.2 Tumor suppressor and oncogene-mediated regulation of (a) glycoly...
Figure 25.3 Schematic representation of biosynthetic pathways diverging from...
Figure 25.4 Alterations in gluconeogenesis and
de novo
serine synthesis in c...
Figure 25.5 Schematic representation of altered glutamine metabolism in canc...
Figure 25.6 Schematic representation of changes in polyamine metabolism in c...
Figure 25.7 Alterations in serine, methionine, and one-carbon metabolism in ...
Figure 25.8 Alterations in branched-chain amino acid metabolism in cancer. M...
Figure 25.9 Metabolic reprogramming supporting antioxidant response machiner...
Figure 25.10 Alterations in (a) purine and (b) pyrimidine nucleotide biosynt...
Figure 25.11 Alterations in lipid metabolism in cancer. Metabolites are show...
Figure 25.12 Niche metabolic alterations in hypoxic tumor core associated wi...
Chapter 26
Scheme 26.1 The pathway of activation of cyclophosphamide and ifosfamide.
Scheme 26.2 Chemical structures of carmustine and lomustine and their degrad...
Figure 26.1 (a) Chemical structures of laromustine and VNP4090CE and (b) Che...
Figure 26.2 Chromatograms from incubation of [14C]laromustine (100 μM) with ...
Figure 26.3 Correlation between the rate of formation of C-7 from [14C]larom...
Figure 26.4 FTMS results for MS/MS of m/z 325 peak using 9.4 T Bruker Qe FTI...
Figure 26.5 Proposed mechanism of formation fragmentation ions of
m/z
251, 1...
Figure 26.6 Proposed fragmentation ions of laromustine of
m/z
325, 327 and 3...
Figure 26.7 Proposed structure and fragmentation ions for C-7.
Figure 26.8 Proposed formation mechanism of MW 214 (C-4 and C-5).
Chapter 27
Figure 27.1 Schematic representation of different types of reactions catalyz...
Figure 27.2 Cross-talk between drug metabolism and endogenous metabolic mach...
Figure 27.3 Role of epigenetics in mediating cross-talk between cancer metab...
Figure 27.4 Role of microenvironment on cross talk between metabolic reprogr...
Chapter 28
Figure 28.1 Delicate balance between optimizing cardiovascular and cancer ou...
Figure 28.2 Cytochrome protein 450 (CYP450) and P-glycoprotein (P-g) in card...
Figure 28.3 Genetic variation and impact on drug response. Variations in pha...
Figure 28.4 Spectrum of phenotypes manifesting from Cytochrome protein 450 (...
Figure 28.5 Genomic, physiologic, and demographic regulators of Cytochrome p...
Figure 28.6 Variations in the genome to the microRNAome can influence the ph...
Figure 28.7 Factors that impact drug response. Overall drug response, and th...
Cover
Title Page
Copyright
Preface
List of Contributors
Table of Contents
Begin Reading
Index
End User License Agreement
iii
iv
xiii
xiv
xv
xvi
xvii
xviii
1
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
66
67
68
69
70
71
72
73
74
75
76
77
78
79
80
81
82
83
84
85
86
87
88
89
90
91
92
93
94
95
96
97
98
99
100
101
103
104
105
106
107
108
109
110
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
126
127
128
129
130
131
132
133
134
135
136
137
138
139
140
141
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
161
162
163
164
165
166
167
168
169
170
171
172
173
174
175
176
177
179
180
181
182
183
184
185
186
187
188
189
190
191
192
193
194
195
196
197
198
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
215
216
217
218
219
220
221
222
223
224
225
226
227
228
229
230
231
232
233
234
235
236
237
238
239
240
241
242
243
244
245
246
247
248
249
250
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
272
273
274
275
276
277
278
279
280
281
282
283
284
285
286
287
288
289
290
291
292
293
294
295
296
297
298
299
300
301
302
303
304
305
306
307
308
309
310
311
312
313
314
315
316
317
318
319
320
321
322
323
324
325
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
342
343
345
346
347
348
349
350
351
352
353
354
355
356
357
358
359
360
361
362
363
364
365
366
367
368
369
370
371
372
373
374
375
376
377
378
379
380
381
382
383
384
385
386
387
388
389
390
391
392
393
394
395
396
397
398
399
400
401
402
403
404
405
407
409
410
411
412
413
414
415
416
417
418
419
420
421
422
423
424
425
427
428
429
430
431
432
433
434
435
436
437
438
439
440
441
442
443
444
445
446
447
448
449
450
451
452
453
454
455
456
457
458
459
460
461
462
463
464
465
466
467
468
469
470
471
472
473
474
475
476
477
478
479
480
481
482
483
484
485
486
487
488
489
490
491
492
493
494
495
496
497
498
499
500
501
502
503
504
505
506
507
508
509
510
511
512
513
514
515
516
517
518
519
520
521
522
523
524
525
526
527
528
529
530
531
532
533
534
535
536
537
538
539
540
541
542
543
544
545
546
547
548
549
550
551
552
553
554
555
556
557
558
559
560
561
562
563
564
565
566
567
568
569
570
571
573
574
575
576
577
578
579
580
581
582
583
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
605
607
608
609
610
611
612
613
614
615
616
617
618
619
620
621
622
623
624
625
626
627
628
629
630
631
632
633
634
635
636
637
638
639
640
641
642
643
644
645
646
647
648
649
650
651
652
653
654
655
656
657
658
659
660
661
662
663
664
665
666
667
668
669
670
671
672
673
674
675
676
677
678
679
680
681
682
683
684
685
686
687
688
689
690
691
692
693
694
695
696
697
698
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
721
722
723
724
725
726
727
728
729
730
731
732
733
734
735
736
737
738
739
740
741
742
743
745
746
747
748
749
750
751
752
753
754
755
756
757
758
759
760
761
762
763
764
765
766
767
768
769
770
771
773
775
776
777
778
779
780
781
782
783
784
785
786
787
788
789
790
791
792
793
794
795
796
797
798
799
800
801
802
803
804
805
806
807
808
809
811
812
813
814
815
816
817
818
819
820
821
822
823
824
825
826
827
828
829
830
831
832
833
834
835
836
837
838
839
841
842
843
844
845
846
847
848
849
850
851
852
853
854
855
856
857
858
859
860
861
862
863
864
865
866
867
868
869
870
871
872
873
874
875
876
877
878
879
880
881
882
883
884
885
886
887
888
889
890
891
892
893
894
895
896
897
898
899
900
901
902
903
904
905
906
907
908
909
910
911
912
913
914
915
916
917
918
919
920
921
922
923
924
925
926
927
928
929
930
931
932
933
934
935
936
937
938
939
940
941
942
943
944
945
946
947
948
949
950
951
952
953
954
955
956
957
958
959
960
961
962
963
964
965
966
967
968
969
970
971
972
973
974
975
976
977
978
979
980
981
982
983
984
985
986
987
988
989
990
991
992
993
994
995
996
997
998
999
1000
1001
1002
1003
1004
1005
1006
1007
1008
1009
1010
1011
1012
1013
1014
Volume 1
Second Edition
Edited by
Ala F. NassarYale University, USA
Associate Editors
Paul F. HollenbergThe University of Michigan Medical, USA
JoAnn ScatinaHealthcare investors, USA
Soumen Kanti MannaSaha Institute of Nuclear Physics, IN
Su ZengZhejiang University, CH
Volume 2
Second Edition
Edited by
Ala F. NassarYale University, USA
Associate Editors
Paul F. HollenbergThe University of Michigan Medical, USA
JoAnn ScatinaHealthcare investors, USA
Soumen Kanti MannaSaha Institute of Nuclear Physics, IN
Su ZengZhejiang University, CH
This second edition first published 2023© 2023 John Wiley & Sons, Inc.
Edition HistoryJohn Wiley & Sons (1e, 2009)
All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, electronic, mechanical, photocopying, recording or otherwise, except as permitted by law. Advice on how to obtain permission to reuse material from this title is available at http://www.wiley.com/go/permissions.
The right of Ala F. Nassar, Paul F. Hollenberg, JoAnn Scatina, Soumen Kanti Manna, and Su Zeng to be identified as the authors of the editorial material in this work has been asserted in accordance with law.
Registered OfficeJohn Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA
For details of our global editorial offices, customer services, and more information about Wiley products visit us at www.wiley.com.
Wiley also publishes its books in a variety of electronic formats and by print-on-demand. Some content that appears in standard print versions of this book may not be available in other formats.
Trademarks: Wiley and the Wiley logo are trademarks or registered trademarks of John Wiley & Sons, Inc. and/or its affiliates in the United States and other countries and may not be used without written permission. All other trademarks are the property of their respective owners. John Wiley & Sons, Inc. is not associated with any product or vendor mentioned in this book.
Limit of Liability/Disclaimer of WarrantyIn view of ongoing research, equipment modifications, changes in governmental regulations, and the constant flow of information relating to the use of medicines, equipment, and devices, the reader is urged to review and evaluate the information provided in the package insert or instructions for each medicine, equipment, or device for, among other things, any changes in the instructions or indication of usage and for added warnings and precautions. While the publisher and authors have used their best efforts in preparing this work, they make no representations or warranties with respect to the accuracy or completeness of the contents of this work and specifically disclaim all warranties, including without limitation any implied warranties of merchantability or fitness for a particular purpose. No warranty may be created or extended by sales representatives, written sales materials or promotional statements for this work. The fact that an organization, website, or product is referred to in this work as a citation and/or potential source of further information does not mean that the publisher and authors endorse the information or services the organization, website, or product may provide or recommendations it may make. This work is sold with the understanding that the publisher is not engaged in rendering professional services. The advice and strategies contained herein may not be suitable for your situation. You should consult with a specialist where appropriate. Further, readers should be aware that websites listed in this work may have changed or disappeared between when this work was written and when it is read. Neither the publisher nor authors shall be liable for any loss of profit or any other commercial damages, including but not limited to special, incidental, consequential, or other damages.
Library of Congress Cataloging-in-Publication Data applied for:
ISBN: 9781119851011[set]
Cover Design: WileyCover Image: © UGREEN 3S/Shutterstock.com
Studies on absorption, distribution, metabolism, and elimination–toxicology (ADME–Tox) have progressed over the years to the point where they now play a major role in drug discovery and development. Until the late 1980s, the primary role of drug metabolism groups in the pharmaceutical industry was to provide ADME–Tox information to support the regulatory package. During the last decade, with the rapid rise in new molecular entities (NMEs) from combinatorial chemistry and high-throughput biological screening, there is an urgent need for the determination of the ADME properties of these NMEs at very early stages in the drug discovery pipeline in order to facilitate the selection of “ideal” drug candidates for further development. Back integration of key studies into the discovery phase has resulted in earlier identification of potential DM/PK and safety liabilities. This information aids in decision making and, in many instances, has been incorporated into criteria for compound advancement into the development phase. Given the need for earlier and more rapid evaluation of a larger number of compounds, drug metabolism scientists have developed and incorporated numerous novel approaches into early drug discovery. These include “humanized” in vitro-based cell systems, sophisticated automation, higher-throughput ADME assays and screens, ultrasensitive analytical technologies, and computational models in order to accelerate the examination of the drug metabolism pathways of their NMEs. The success of this approach is evident, as the number of failures due to DM/PK liabilities has dramatically decreased. Clearly, there is a growing need for improving and expanding education of students as well as current practitioners involved in investigations in these areas. The needs for continuing education in the rapidly expanding and dynamic area of ADME-Tox studies are not being met at the university level, and oftentimes this is being taught in a piece-meal fashion on the job in pharmaceutical industries.
Therefore, the goal of this book is to provide a systematic approach for the education of students at the university level, as well as younger researchers and scientists changing fields in the pharmaceutical industry to improve their knowledge of drug metabolism by presenting in-depth coverage of the drug disposition process, pharmacokinetic drug–drug interactions, theory, and evaluation approaches and improving the decision-making process used for the structural modification of drug candidates to reduce toxicity. ADME-Tox experts in the field from both industry and academia have joined forces and offered their time to write this book, introducing students to modern concepts and practices of ADME-Tox. This book provides basic training in the areas of drug metabolism and disposition, including training programs for students as well as new employees in the pharmaceutical industry. Mastery of the material in this text will allow them to apply state-of-the-art research tools to in vitro and in vivo metabolism studies and contribute greatly to their abilities to perform pharmaceutical research in support of industrial, academic, and regulatory agency needs. One emphasis of the current edition – as evident from the title – is to improve our understanding of drug metabolism and discovery in the context of cancer. Metabolic reprogramming, which is a hallmark of cancer, is a target of several existing and experimental drugs. This book will discuss the nature of metabolic reprogramming in cancer and its cross-talk with drug metabolism. It will also present strategies and tools for elucidation of heterogeneity in drug disposition and metabolism in tumors as well as for therapeutic personalization.
This textbook consists of five parts. Part I provides an introduction to drug metabolism. Part II presents the in vitro and in vivo technologies used to investigate the metabolism of drugs and drug candidates. Part III presents an important area of drug–drug interaction. Part IV discusses the toxicity of drugs and their metabolites. Part V of this volume provides an up-to-date series of chapters on the applications of drug metabolism in cancer. This book provides a unique and useful approach for all those involved in drug discovery and development, and for clinicians and researchers in drug metabolism, pharmacology, and clinical pharmacology.
The editors and contributing authors greatly appreciate the commitment of the publishers to make this book available to our scientific colleagues in developing countries to enhance their knowledge in the area of ADME-Tox and to help them in furthering their careers in this very important area of research. Finally, we thank our many colleagues worldwide who have contributed to the development of the knowledge and techniques described in this book. We feel very fortunate to be able to participate in an area of scientific pursuit in which cooperation and collaboration between investigators in industry, academic institutions, and regulatory agencies is so strongly encouraged and highly valued. Drug Metabolism Handbook is a comprehensive reference devoted to the current state of research on the impact of various disease states on drug metabolism. The book contains valuable insights into mechanistic effects and examples of how to accurately predict drug metabolism during these different pathophysiological states. Each chapter clearly presents the effects of changes in drug metabolism enzymes and drug transporters on the pharmacokinetics, disposition, and potential toxicity of NMEs.
ALA F. NASSAR
School of Medicine, Yale University, CT, USA
August 2022
Shibdas Banerjee, Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati, India
Ganesh K. Barik, Molecular Oncology Laboratory, National Centre for Cell Science, Pune, Maharashtra, India
and
Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
Craig Beavers, Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY, USA
Praneeta P. Bhavsar, Proteomics Lab, National Centre for Cell Science, Pune, Maharashtra, India
and
Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
Sherry-Ann Brown, Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
Joe R. Cannon, Bristol Myers Squibb, Lawrenceville, NJ, USA
Purva S. Damale, Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati, India
Carl Davis, Pharmacokinetics, Dynamics & Metabolism, Pfizer Inc., La Jolla, CA, USA
Gift Echefu, Department of Medicine, Baton Rouge General Medical Center, Baton Rouge, LA, USA
Olubadewa Fatunde, Division of Cardiology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
Mark P. Grillo, Cytokinetics, South San Francisco, CA, USA
Umesh M. Hanumegowda, ViiV Healthcare, Branford, CT, USA
Paul F. Hollenberg, Department of Pharmacology, The University of Michigan Medical School, Ann Arbor, MI, USA
Eli G. Hvastkovs, Department of Chemistry, East Carolina University, Greenville, NC, USA
Sun Min Jung, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
Bhargab Kalita, Proteomics Lab, National Centre for Cell Science, Pune, Maharashtra, India
and
Amrita School of Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Kochi, Kerala, India
Sailaja Kamaraju, Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
Roberta S. King, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
Ivan King, Department of Clinical Operations, Hinova Pharmaceuticals Inc., San Diego, CA, USA
Lawrence H. Lash, Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
Jiapeng Li, Department of Clinical Pharmacy, University of Michigan, Ann Arbor, MI, USA
Subhabrata Majumder, Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
and
Homi Bhabha National Institute, Mumbai, India
Soumen Kanti Manna, Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
and
Homi Bhabha National Institute, Mumbai, India
Meera Mohan, Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
Ala F. Nassar, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
Carolyn Oxencis, Department of Pharmacy, Froedtert and the Medical College of Wisconsin, Milwaukee, WI, USA
Debasish Prusty, Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
and
Homi Bhabha National Institute, Mumbai, India
Srikanth Rapole, Proteomics Lab, National Centre for Cell Science, Pune, Maharashtra, India
Dan A. Rock, Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, San Francisco, CA, USA
James F. Rusling, Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, USA
and
Department of Surgery, Uconn Health Center, Farmington, CT, USA
and
School of Chemistry, National University of Ireland, Arts and Science Building, University Rd, Galway, Ireland
Manas K. Santra, Molecular Oncology Laboratory, National Centre for Cell Science, Pune, Maharashtra, India
Tanisha Sharma, Molecular Oncology Laboratory, National Centre for Cell Science, Pune, Maharashtra, India
and
Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
Thomas R. Sharp, Department of Chemistry and Chemical Engineering, Tagliatela College of Engineering, University of New Haven, West Haven, CT, USA
Logan S. Smith, Department of Clinical Pharmacy, University of Michigan, Ann Arbor, MI, USA
Khushman Taunk, Proteomics Lab, National Centre for Cell Science, Pune, Maharashtra, India
and
Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, Nadia, West Bengal, India
Prakash Vachaspati, The Biocon Bristol Myers Squibb Research & Development Center, Bangalore, India
Brianna Wallace, School of Pharmacy, Medical College of Wisconsin, Milwaukee, WI, USA
Hongbing Wang, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
Jiaqi Wang, Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Cancer Center, Zhejiang University, Hangzhou, China
and
Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China
Yingying Wang, Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Cancer Center, Zhejiang University, Hangzhou, China
and
Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China
Yu Wang, Institute of Drug Metabolism