دسترسی نامحدود
برای کاربرانی که ثبت نام کرده اند
برای ارتباط با ما می توانید از طریق شماره موبایل زیر از طریق تماس و پیامک با ما در ارتباط باشید
در صورت عدم پاسخ گویی از طریق پیامک با پشتیبان در ارتباط باشید
برای کاربرانی که ثبت نام کرده اند
درصورت عدم همخوانی توضیحات با کتاب
از ساعت 7 صبح تا 10 شب
ویرایش: 2
نویسندگان: Shuguang Ma (editor). Swapan Chowdhury (editor)
سری:
ISBN (شابک) : 0128200189, 9780128200186
ناشر: Elsevier Science
سال نشر: 2020
تعداد صفحات: 684
زبان: English
فرمت فایل : PDF (درصورت درخواست کاربر به PDF، EPUB یا AZW3 تبدیل می شود)
حجم فایل: 24 مگابایت
در صورت تبدیل فایل کتاب Identification and Quantification of Drugs, Metabolites, Drug Metabolizing Enzymes, and Transporters: Concepts, Methods and Translational Sciences به فرمت های PDF، EPUB، AZW3، MOBI و یا DJVU می توانید به پشتیبان اطلاع دهید تا فایل مورد نظر را تبدیل نمایند.
توجه داشته باشید کتاب شناسایی و تعیین مقدار داروها ، متابولیت ها ، آنزیم های متابولیزه دارو و ناقلین: مفاهیم ، روش ها و علوم ترجمه ای نسخه زبان اصلی می باشد و کتاب ترجمه شده به فارسی نمی باشد. وبسایت اینترنشنال لایبرری ارائه دهنده کتاب های زبان اصلی می باشد و هیچ گونه کتاب ترجمه شده یا نوشته شده به فارسی را ارائه نمی دهد.
شناسایی و کمیت داروها، متابولیتها، آنزیمهای متابولیزهکننده دارو، و انتقالدهندهها، ویرایش دوم، کاملاً بهروز شده است تا نمای کلی از پیشرفتهای متعدد دهه گذشته در فناوریهای تحلیلی برای تشخیص و تعیین کمیت داروها ارائه کند. متابولیت ها و نشانگرهای زیستی این نسخه جدید فراتر از LC-MS است و دارای فصول کاملاً جدیدی در مورد نحوه ارزیابی جذب، توزیع، متابولیسم و دفع دارو، پتانسیل سمیت کبدی و کلیوی، ایمنی زایی بیوتراپی ها و ابزارهای ترجمه برای پیش بینی دوز انسانی، ایمنی و اثربخشی دارو است. مولکول های کوچک و بیولوژیک ها این کتاب یک کتاب راهنما و مرجع مهم برای فارماکولوژیست ها، سم شناسان، دانشمندان بالینی و دانشجویان علاقه مند به رشته های فارماکولوژی، بیوشیمی و متابولیسم داروها خواهد بود.
Identification and Quantification of Drugs, Metabolites, Drug Metabolizing Enzymes, and Transporters, Second Edition, is completely updated to provide an overview of the last decade’s numerous advances in analytical technologies for detection and quantification of drugs, metabolites, and biomarkers. This new edition goes beyond LC-MS and features all-new chapters on how to evaluate drug absorption, distribution, metabolism, and excretion, potential for hepatic and renal toxicity, immunogenicity of biotherapeutics and translational tools for predicting human dosage, safety and efficacy of small molecules and biologics. This book will be an important handbook and desk reference for pharmacologists, toxicologists, clinical scientists, and students interested in the fields of pharmacology, biochemistry, and drug metabolism.
Cover IDENTIFICATION AND QUANTIFICATION OF DRUGS, METABOLITES, DRUG METABOLIZING ENZYMES, AND TRANSPORTERS: Concepts, Methods, and Translational Sciences Copyright Contributors Foreword References Preface Part I: Techniques for identifying and quantifying drugs and metabolites Bioanalysis of small and large molecule drugs, metabolites, and biomarkers by LC-MS Introduction Complexity of contemporary bioanalysis Bioanalytical requirements for supporting discovery, nonclinical, and clinical studies Current regulatory landscape for bioanalysis General considerations for bioanalysis for sample collection Diagnosis and mitigation of nonspecific adsorption loss for urine bioanalysis Tissue bioanalysis Managing unstable metabolites such as acyl glucuronide General considerations for bioanalysis for extraction, chromatography, and MS detection Selected applications for LC-MS bioanalysis LC-MS of large molecules LC-MS bioanalysis of mAb LC-MS bioanalysis of ADC LC-MS bioanalysis of PDC LC-MS bioanalysis of protein biomarkers LC-MS bioanalysis of drug metabolizing enzymes and transporters LC-MS bioanalysis of half-life extended biotherapeutics LC-MS bioanalysis using microsampling Microsampling sample collection in bioanalysis Microsampling in blood and plasma Bioanalytical method development for microsampling in blood and plasma Biomarkers quantitation Leukotriene B4 (authentic matrix and authentic analyte) 4β-Hydroxylcholesterol and 7α-hydroxy-4-cholesten-3-one (C4) (surrogate matrix and authentic analyte) Fatty acid amide hydrolase biomarkers (authentic matrix and surrogate analytes) LC-MS analysis of metabolites and MIST Metabolite quantitation strategy LC-MS bioanalysis of polar metabolites Bioanalysis of chiral compounds Unexpected assay challenge during analysis of patient samples Conclusion and future perspective References Recent advances in mass spectrometric and other analytical techniques for the identification of drug metabolites Introduction Sample preparation strategies Solid-phase extraction Supported phase liquid extraction Liquid-liquid extraction Protein precipitation Optical detectors and chromatographic separation techniques Ultra-performance liquid chromatography Supercritical fluid chromatography Different types of Ionization techniques and mass spectrometric scan functions Ionization techniques Mass analyzers and different acquisition modes Inductively coupled mass spectrometry (ICP-MS) Wet chemistry techniques combined with MS Hydrogen/deuterium (H/D) exchange Chemical derivatization Increased LC retention and sensitivity in LC-MS for polar metabolites Unusual metabolite characterization Unstable metabolites Isomeric metabolite characterization Determination of site of glucuronidation Nuclear magnetic resonance spectroscopy Conclusion and future trends References High-resolution mass spectrometry-based data acquisition and data-mining technologies for Introduction HRMS-based data acquisition technologies for metabolite identification Data-dependent MS/MS acquisition Ion intensity-dependent acquisition Accurate mass inclusion list-dependent acquisition Mass defect-dependent acquisition Isotope pattern-dependent acquisition Pseudo-neutral loss-dependent acquisition Background-exclusion data-dependent acquisition Data-independent acquisition MSE Sequential windowed acquisition of all theoretical fragment ions (SWATH) All ion fragmentation HRMS-based data-processing techniques for metabolite identification Targeted data-mining technology Extracted ion chromatography Mass defect filter Isotope pattern filter Product ion filter and neutral loss filter Nontargeted data-processing approaches Background subtraction Metabolomics approach Software-assisted metabolite prediction and identification Software for site of metabolism prediction Software for metabolite structural identification Applications of HRMS technologies in metabolite identification experiments Metabolic soft-spot analysis Reactive metabolite screening In vivo metabolite profiling and identification Detection and structural characterization of traditional Chinese medicine components in biological systems HRMS-based data acquisition techniques for profiling and characterizing TCM components in biological samples Targeted data-mining techniques applied to detection and identification of in vivo TCM components Untargeted data-mining techniques for detection and characterization of TCM components LC-HRMS-based techniques for determining metabolic pathways of individual TCM components A comprehensive analytical strategy for study of exposure, metabolism, and disposition of TCM components Conclusion and future perspectives Acknowledgments References Methods for metabolite generation and characterization by NMR Introduction Methods for scaled-up production of drug metabolites Chemical synthesis Synthesis of phase I oxidized metabolites Example: Synthesis of multiple oxidized metabolites for bioactivity testing Example: Synthetic routes to active metabolites of clopidogrel Synthesis of phase II conjugated metabolites Biomimetic chemistry Electrochemistry Example: Production of CYP1A1 and CYP1B1-mediated amodiaquine metabolite M2 by electrochemistry Mammalian tissue fractions Example: Species-dependent regioselective hydroxylation of drugs Whole-cell microbial biotransformation Example: Accessing disproportionate human metabolites for toxicology studies Example: Accessing multipathway-derived metabolites Recombinant enzymes Cytochrome P450 enzymes Non-CYP phase I enzymes Use of other oxidizing enzymes Recombinant UGTs Combined synthetic and biosynthetic approach Example: Accessing human metabolites of drugs subject to metabolic shunting Purification and structure elucidation of metabolites Metabolite purification Example: Purification of metabolites from excreta for use as analytical standards Metabolite structure elucidation using NMR spectroscopy 1D and 2D NMR experiments for metabolite structure elucidation Stepwise determination of metabolite structures Conclusions and future direction Acknowledgments References Application of SFC for bioanalysis Introduction Considerations for SFC method development Detection in SFC Sample preparation Examples Drugs of abuse, doping control, and toxicological analysis Pharmaceuticals and clinical analysis Endogenous metabolites, metabolomics, and lipidomics Conclusion References AMS in drug development: Exploring the current utility of AMS and future opportunities for absolute bioavailab ... Introduction Introduction of the AMS technique AMS vs. conventional methods Application of AMS to drug development Sample preparation (``graphitization´´) Analysis by AMS Data acquisition and calculations Calculation of analyte concentration Validation of LC+AMS Evolution of AMS Clinical study design definitions First drug development clinical study application of AMS Hybrid studies (macrotracer) Low tracer dose studies Concomitant microtracer: Design and delivery Absolute bioavailability assessments using AMS Improved efficiencies in drug development Conclusions Future perspectives References Part II: Drug metabolism enzymes, transporters and drug-drug interaction Using in vitro methods to determine P450s responsible for metabolism and discrimination from other oxidative p ... Introduction Recombinant P450 assays Human liver microsome-based methods Antibodies Chemical inhibitors Silensomes Correlation analysis P450 vs FMO/AO metabolism Determination of fmCYP3A Current methods for fmCYP3A Regulatory guidance/risk assessment/examples Conclusion and future directions References Evaluation of the clearance mechanism of non-CYP-mediated drug metabolism and DDI as a victim drug Introduction UDP-glucuronosyltransferase (UGT) Reaction phenotyping of UGT enzyme(s) Incubation with pooled HLM Incubation with recombinant human UGT enzymes Inhibition study with chemical inhibitors of UGTs Challenges Clinical relevance and drug-drug interactions (DDI) Flavin monooxygenase (FMO) The FMO catalytic cycle Determination of in vitro relative contribution of FMO vs CYP Experimental design for FMO phenotyping Challenges Clinical significance and DDI Monoamine oxidase (MAO) Experimental designs for MAO phenotyping Challenges Clinical significance Aldehyde oxidase Experimental designs for AOX1 reaction phenotyping Challenges Clinical significance and DDI Xanthine oxidase (XO) Experimental designs for XO phenotyping Challenges Clinical significance and DDI Carboxylesterases (CES) Experimental design for CES reaction phenotyping Challenges Clinical significance: CES and DDI Aldo-keto reductase (AKR) Experimental design for AKR1C reaction phenotyping AKR: Human-animal comparison Challenges Clinical significance Future trends Conclusion Acknowledgment References In vitro characterization and in vitro to in vivo predictions of drug-drug interactions Introduction In vitro assessment of metabolism-based drug interaction potential Characterization of the substrate Characterization of the inhibitor Reversible inhibitors Time-dependent inhibitors Enzyme induction Quantitative in vitro to in vivo predictions Basic principles of reversible inhibition predictions Selection of inhibitor concentration, [I] Prediction of irreversible inhibition Prediction of drug metabolizing enzyme induction Inhibition and induction of first-pass intestinal metabolism and integration of multiple DDI mechanisms Clinical drug interaction assessment Conclusions References Role of transporters in drug disposition and drug-drug interactions Introduction Overview of membrane transporters The ATP-binding cassette superfamily P-glycoprotein BCRP Multidrug resistance proteins Bile salt export protein The solute carrier protein superfamily Organic anion transporting polypeptides Organic anion transporters Organic cation transporters Multidrug and toxin extrusion transporters Clinical significance of transporter-mediated drug disposition and drug-drug interactions Tools to assess transporter liabilities in drug discovery and development Recombinant transfected cell-based systems Membrane vesicle-based assay Polarized cell-based systems and bidirectional transporter assays Primary cell-based assays The use of animal models to assess transporter liabilities in drug development In silico modeling of transporter proteins Application of biomarkers for in vivo assessment of drug transporter activity Regulatory landscape of evaluating transporter-mediated drug interactions Determining if the investigational drug is a substrate of transporters Determining if the investigational drug is an inhibitor of transporters Challenges and perspectives on transporter-mediated drug interactions References Mechanisms and clinical relevance of pharmacokinetic-based clinical drug-drug interactions for drugs r Introduction Enzyme-mediated DDIs NMEs as substrates of enzymes NMEs as inhibitors of enzymes NMEs as inducers of enzymes Transporter-mediated DDIs NMEs as substrates of transporters NMEs as inhibitors of transporters PBPK modeling and simulations in DDI prediction PGx studies Other mechanisms: Absorption-based DDIs Conclusion References Quantifying drug metabolizing enzymes and transporters by LC-MS/MS proteomics Introduction Basic workflow of DMET quantitative proteomics Selection of proteotypic peptides Sample procurement, homogenization, and protein extraction Protein extraction and digestion methods Post-digestion processing Peptide separation using liquid chromatography (LC) Mass spectrometry (MS) analysis of peptide signal Untargeted MS acquisition approaches Protein quantification approaches Factors affecting DMET protein quantification Optimized quantitative analysis approaches Use of multiple peptides Use of multiple product ions Inclusion of a positive control sample Calibration curve, surrogate matrix, and LLOQ SIL peptide and exogenous protein internal standards Optimized practices in targeted quantitative proteomics Applications of quantitative DMET proteomics Characterization of in vitro and in vivo models In vitro to in vivo extrapolation (IVIVE) of drug clearance Interspecies differences in protein expression Differential tissue and regional protein distribution Subcellular localization of proteins Interindividual variability and precision medicine Drug-drug interaction (DDI) potential (induction/suppression) Drug/metabolite-protein interactions Conclusion References Protein drug-drug interactions for therapeutic modalities Introduction DDI mechanisms Major mechanisms of TP-DDI In vitro effects on CYP enzymes and transporters TP-DDI observed in clinical studies Cytokine-dependent interactions Cytokines and therapeutic proteins targeting cytokines Immunomodulatory therapeutic proteins Immunogenicity-dependent interactions Target-dependent interactions Other interactions Interactions based on physiology Interactions based on binding to proteoglycans Interactions of antibody-drug conjugates Potential DDI between emerging modalities Oligonucleotide and mRNA-based drugs Cell-based therapies Oncolytic viruses Immunocytokines Risk assessment and strategies to evaluate potential TP-DDI Risk assessment Exploratory studies to assess TP-DDI risk Dedicated TP-DDI studies Design of studies Specific study considerations Conclusion and future perspectives References Part III: Strategy related to drug metabolism and safety Metabolites in safety testing (MIST) Introduction History of guidance on safety testing of drug metabolites Summary of the metabolites in safety testing (MIST) guidance Technological approaches for MIST assessment High-performance liquid chromatography separation and radiometric detection High-resolution mass spectrometry Nuclear magnetic resonance spectroscopy Accelerator mass spectrometry and cavity ring-down spectroscopy Post-acquisition software tools for metabolite identification Semiquantitative and quantitative assessment of metabolite coverage Calibrator approaches Mixed-matrix approach for direct comparison of metabolite levels in human and animal plasma samples and AUC pooling Bioanalytical methods A typical MIST strategy Stage 1. Before entering human studies Stage 2. During Phase 1 human clinical studies Stage 3. Before the start of Phase 3 human clinical studies Metabolite safety assessment beyond the MIST guidance documents Conclusion and future outlook References The use of stable isotopes in drug metabolism studies Introduction Use of stable labels for metabolite detection and identification Stable labels and their application Beyond typical choices of isotopes Types of studies in drug metabolism Cases studies Vismodegib and oxidative pyridine ring cleavage Using H218O and D2O to understand tofacitinib metabolism Stable-labeled glutathione as a trapping agent for detection of reactive metabolites A disconnect between endogenous and [13C]-labeled niacin Deuterated drugs Deuterium in drug design Relevant drug-metabolizing enzymes for utilizing kinetic isotope effects Cytochrome P450 metabolism Monoamine oxidase metabolism Aldehyde oxidase metabolism Case studies: Deuterated versions of old drugs Lowering clearance Improving bioavailability Mitigating reactive metabolite formation/drug-drug interaction Slowing chiral inversion How deuterium should be assessed in drug discovery? Conclusions and future perspectives References Assessment of stereoselectivity in pharmacology, toxicology, and drug metabolism Introduction Regulatory considerations on developing chiral drugs Stereoselectivity in pharmacodynamics Selective competitive antagonism Enantiomers have the opposite effects One enantiomer has side effects Different biological activities Complementarity of enantiomeric effects The biological activity of a drug is produced by single enantiomer Different action targets present different characteristics Same biological activity Stereoselectivity in pharmacokinetics and ADME properties Stereoselectivity of chiral drug absorption and transport Stereoselectivity of chiral drug distribution Stereoselectivity of interaction between chiral drugs and plasma proteins Stereoselectivity of interaction between chiral drugs and tissues Chirality in drug metabolism Stereoselectivity of chiral drug excretion Methods for studying chiral drug interactions Stereoselectivity in toxicity Toxicity of chiral drugs Enantiomeric biotransformation increases toxicity Chiral inversion increases toxicity Toxicity or adverse reactions are inseparable from pharmacological activity Active enantiomers are potentially toxic Chiral inversion mechanisms Chiral metabolic inversion of nonsteroidal anti-inflammatory drugs Inversion of the opposite metabolic pathway Stereoselective analytical methods High-performance liquid chromatography Chiral stationary phases Chiral mobile phase additives Chiral derivatization reagents Gas chromatography Chiral stationary phases Chiral derivatization reagents Supercritical fluid chromatography Capillary electrophoresis Immunoassay Conclusion References Progress of derisking strategies for drug-induced liver injury (DILI) in the last two decades Introduction Challenges in predicting DILI Bioactivation of drugs to reactive intermediates Metabolite identification via trapping studies Covalent binding studies in vitro and in vivo Reactive phase II conjugates of carboxylic acid-containing molecules Glaxo Smith Kline multi-assay approach to assess DILI risk from bioactivation Transcriptomics approach Immunomics approach Mitochondrial impairment Transporter inhibition Overarching derisking approaches independent of mechanism Dose, exposure, and physiochemical properties Multiparametric approaches European Federation of Pharmaceutical Industries and Associations (EFPIA) three-tiered roadmap Roche approach using a combination of assays Astra Zeneca integrated in vitro hazard matrix and Bayesian machine learning approach Pfizer hepatic risk matrix (HRM) approach Summary References Predictive and translational models for renal drug safety evaluation Background and introduction 2D in vitro models for nephrotoxicity screening Emerging models for renal safety screening Translatable kidney safety biomarkers Context of use of in vitro PTEC models-Mechanistic vs. predictive Outlook and future perspectives References Immunogenicity: An introduction to its role in the safety and efficacy of biotherapeutics Introduction Overview of immunogenicity Overview of immune response mechanisms Humoral immunogenicity Cellular immunogenicity Presentation via MHC I on the cell surface and binding to CD8+ T cells Activation and cellular response of CD8+ T cells Humoral immunogenicity: Overall risk assessment and mitigation strategies Case studies Case study 1: PK assessment in the presence of ADA in animal models Case study 2: Immune response to AAV vectors in animal models Case study 3: In silico immunogenicity prediction Cellular immunogenicity risk assessments In silico prediction tool ELISpot Chromium 51 Flow cytometry In vivo analysis Mitigation strategy and case studies for cellular immunogenicity Modification of AAV Case study: AAV2 YF engineered AAV2 capsid Humanization of CAR construct Case study: Reducing the immunogenicity of CAR construct Lymphodepletion Chapter summary Acknowledgments References Part IV: Translational sciences Application of genetically modified rodent models in drug discovery and development for translation of clinical ADME properties Introduction The rise of novel animal models in drug research Knockout animal models Xenobiotic receptor KO models Advantages Disadvantages Humanized transgenic animal models Utility of humanized CYP mouse models to understand human ADME Humanized liver chimeric mouse models Advantages Disadvantages Conclusions and future perspectives References Advances in CRISPR technologies enable novel in vitro tools for ADME studies Introduction In vitro applications of CRISPR Genome editing with CRISPR Repression of target genes with CRISPR Activation of target genes with CRISPR Genetic screens with CRISPR Examples of CRISPR application in ADME studies in vitro Genetic modification of CYP3A5*3 in Huh-7 cells Endogenous Canine P-gp knockout in Madin-Darby canine kidney cells Potential opportunities and limitations for CRISPR applications in ADME studies in vitro Limitations of CRISPR in ADME studies in vitro Potential opportunities for CRISPR applications in ADME studies in vitro References In vitro-in vivo extrapolation of human hepatic and renal clearance Introduction ECCS framework to identify rate-determining process for CL Current IVIVE approaches to predict clearance Hepatic clearance Metabolic clearance and in vitro methodologies IVIVE of metabolic clearance In vitro-to-in vivo correlation (IVIVC) of metabolic clearance Transporter and metabolism interplay In vitro tools and IVIVE of overall hepatic clearance IVIVC of overall hepatic clearance Renal clearance Metabolic clearance and IVIVE IVIVE of transporter-mediated CL and tubular reabsorption Conclusions References The role of quantitative modeling and simulation in translational science Introduction Pharmacokinetic modeling Pharmacokinetic modeling for small molecule drugs Compartmental and population-based PK models Physiologically-based pharmacokinetic modeling Physiological-based absorption modeling to assess food's and pH's effects on pharmacokinetics Modeling the interplay between gut transporters and CYP3A Distribution modeling using PBPK Prediction of human clearance (hepatic, renal, and biliary excretion) Gaps and challenges in PBPK modeling Pharmacokinetic modeling for biologics Modeling the absorption and distribution of biologics Target-mediated drug disposition models and their role in understanding clearance of biologics General dynamic of TMDD model Constant receptor amount (Rtotal) model Quasi-equilibrium model (QE) Quasi-steady-state model Michaelis-Menten model Modeling pharmacodynamic response Pharmacodynamic modeling for small molecule drugs PKPD modeling to predict efficacy of D2 receptor antagonists for the treatment of schizophrenia Using translational PKPD modeling to accelerate EGFR inhibitor development for the treatment of lung cancer Special considerations for biologic pharmacodynamic modeling Conclusions References PK/PD-driven starting and effective human dose determination for immuno-oncology drugs Introduction Dose selection for immune-activating agents Dose selection for T-cell engaging bispecific molecules Dose selection for emerging class of I-O therapies Future directions References Index Back Cover