An Introduction to Medicinal Chemistry
Námskeið
- LYF302F Lyfjaefnafræði/Lyfjahönnun.
Ensk lýsing:
For many people, taking some form of medication is part of everyday life, whether for mild or severe illness, acute or chronic disease, to target infection or to relieve pain. However for most it remains a mystery as to what happens once the drug has been taken into the body: how do the drugs actually work? Furthermore, by what processes are new drugs discovered and brought to market? An Introduction to Medicinal Chemistry, sixth edition, provides an accessible and comprehensive account of this fascinating multidisciplinary field.
Assuming little prior knowledge, the text is ideal for those studying the subject for the first time. Part one of the book introduces the principles of drug action via targets such as receptors and enzymes. The book goes on to explore how drugs work at the molecular level (pharmacodynamics), and the processes involved in ensuring a drug meets its target (pharmacokinetics). Further sections cover the processes by which drugs are discovered and designed, and what has to happen before a drug can be made available to the public.
The book concludes with a selection of current topics in medicinal chemistry, and a discussion of various key drug groups. The subject is brought to life throughout by engaging case studies highlighting particular drugs and the stories behind their discovery and development. The Online Resource Centre features: For students: · Multiple Choice Questions to support self-directed learning · Web articles describing recent developments in the field and further information on topics covered in the book · Journal Club to encourage students to critically analyse the research literature · Molecular Modelling Exercises, with new exercises in Chem3D · New assignments to help students develop data analysis and problem solving skills For registered adopters of the book: · A test bank of additional multiple-choice questions, with links to relevant sections in the book · Answers to end-of-chapter questions.
Lýsing:
Medication is widely used to support the human body to fight against infection and pain. In an era of pharmaceutical and medicinal challenges, we have all become more familiar with drug production and distribution. However, do we really know what happens before those drugs are distributed? What's the process behind drug discovery? How do our bodies interact with those chemicals? An Introduction to Medicinal Chemistry, 7th edition, offers a complete and accessible approach to this multidisciplinary field.
Its student-friendly writing style makes this text an ideal tool for those coming to the subject for first time, but also for students looking to deepen their understanding. The book guides students through understanding the principles of drug action targets in Part A, to how drugs interact at a molecular level with our organs to offer therapeutic value in Part B, and exploring drug design and discovery, as well as regulatory procedures in Part C.
Offering a practical approach, Part D provides a deeper look at specific tools and techniques of medicinal chemistry, concluding with emerging topics including antibodies and anticancer agents in Part E. From principles to practice, accompanied by examples and case studies emerging from current biomedical research, the book will equip students with a robust understanding of medicinal chemistry, to prepare them for future success.
Annað
- Höfundur: Graham L. Patrick
- Útgáfa:7
- Útgáfudagur: 2023-04-06
- Engar takmarkanir á útprentun
- Engar takmarkanir afritun
- Format:ePub
- ISBN 13: 9780192636478
- Print ISBN: 9780198866664
- ISBN 10: 0192636472
Efnisyfirlit
- Cover
- Title Page
- Copyright page
- Preface
- About the Book
- Emboldened key words
- Boxes
- Key points
- Struggle Alert
- Questions
- Further reading
- Appendix and glossary
- Links
- Disclaimer
- About Oxford Learning Link
- Student resources
- Multiple choice questions
- Rotatable 3D structures
- Web articles
- Molecular modelling
- Protein Data Bank
- Lecturer resources
- Test Bank
- Answers
- Figures from the book
- PowerPoint slides
- Student resources
- General interest
- Synthesis
- Clinical correlation
- 1.1 What is a drug?
- 1.2 Drug targets
- 1.2.1 Cell structure
- 1.2.2 Drug targets at the molecular level
- 1.3 Intermolecular bonding forces
- 1.3.1 Electrostatic or ionic bonds
- 1.3.2 Hydrogen bonds
- 1.3.2.1 Conventional hydrogen bonds
- 1.3.2.2 Unconventional hydrogen bonds
- 1.3.3 Van der Waals interactions
- 1.3.4 Dipole–dipole, ion–dipole, and cation–π interactions
- 1.3.5 π–π interactions
- 1.3.6 Halogen bonds
- 1.3.7 Repulsive interactions
- 1.3.8 The role of water and hydrophobic interactions
- 1.4 Pharmacokinetic issues and medicines
- 1.5 Classification of drugs
- 1.5.1 By pharmacological effect
- 1.5.2 By chemical structure
- 1.5.3 By target system
- 1.5.4 By target molecule
- 1.6 Naming of drugs and medicines
- Questions
- Further Reading
- Websites
- List of Key Terms
- 2 Protein structure and function
- 2.1 The primary structure of proteins
- 2.2 The secondary structure of proteins
- 2.2.1 The α-helix
- 2.2.2 The β-pleated sheet
- 2.2.3 The β-turn
- 2.3 The tertiary structure of proteins
- 2.3.1 Covalent bonds: disulphide links
- 2.3.2 Ionic or electrostatic bonds
- 2.3.3 Hydrogen bonds
- 2.3.4 Van der Waals and hydrophobic interactions
- 2.3.5 Relative importance of bonding interactions
- 2.3.6 Role of the planar peptide bond
- 2.4 The quaternary structure of proteins
- 2.5 Translation and post-translational modifications
- 2.6 Proteomics
- 2.7 Protein function
- 2.7.1 Structural proteins
- 2.7.2 Transport proteins
- 2.7.3 Enzymes and receptors
- 2.7.4 Miscellaneous proteins and protein–protein interactions
- Questions
- Oxford Learning Link
- Further Reading
- List of Key Terms
- 3 Enzymes: structure and function
- 3.1 Enzymes as catalysts
- 3.2 How do enzymes catalyse reactions?
- 3.3 The active site of an enzyme
- 3.4 Substrate binding at an active site
- 3.5 The catalytic role of enzymes
- 3.5.1 Binding interactions
- 3.5.2 Acid/base catalysis
- 3.5.3 Nucleophilic groups
- 3.5.4 Stabilization of the transition state
- 3.5.5 Cofactors
- 3.5.6 Naming and classification of enzymes
- 3.5.7 Genetic polymorphism and enzymes
- 3.6 Regulation of enzymes
- 3.7 Isozymes
- 3.8 Enzyme kinetics
- 3.8.1 The Michaelis–Menten equation
- 3.8.2 Lineweaver–Burk plots
- Questions
- Further Reading
- List of Key Terms
- 4 Receptors: structure and function
- 4.1 Role of the receptor
- 4.2 Neurotransmitters and hormones
- 4.3 Receptor types and subtypes
- 4.4 Receptor activation
- 4.5 How does the binding site change shape?
- 4.6 Ion channel receptors
- 4.6.1 General principles
- 4.6.2 Structure
- 4.6.3 Gating
- 4.6.4 Ligand-gated and voltage-gated ion channels
- 4.7 G-Protein-coupled receptors
- 4.7.1 General principles
- 4.7.2 Structure of G-protein-coupled receptors
- 4.7.3 The rhodopsin-like family of G-protein-coupled receptors
- 4.7.4 Dimerization of G-coupled receptors
- 4.8 Kinase receptors
- 4.8.1 General principles
- 4.8.2 Structure of tyrosine kinase receptors
- 4.8.3 Activation mechanism for tyrosine kinase receptors
- 4.8.4 Tyrosine kinase receptors as targets in drug discovery
- 4.8.4.1 The ErbB family of tyrosine kinase receptors
- 4.8.4.2 Vascular endothelial growth factor receptors
- 4.8.4.3 Platelet-derived growth factor receptors
- 4.8.4.4 Stem cell growth factor receptors
- 4.8.4.5 Anaplastic lymphoma kinase
- 4.8.4.6 The RET receptor
- 4.8.4.7 Hepatocyte growth factor receptor or c-MET receptor
- 4.9 Intracellular receptors
- 4.10 Regulation of receptor activity
- 4.11 Genetic polymorphism and receptors
- Questions
- Further Reading
- List of Key Terms
- 5.1 Signal transduction pathways for G-protein-coupled receptors (GPCRs)
- 5.1.1 Interaction of the receptor–ligand complex with G-proteins
- 5.1.2 Signal transduction pathways involving the α-subunit
- 5.2 Signal transduction involving G-proteins and adenylate cyclase
- 5.2.1 Activation of adenylate cyclase by the αs-subunit
- 5.2.2 Activation of protein kinase A
- 5.2.3 The Gi-protein
- 5.2.4 General points about the signalling cascade involving cyclic AMP
- 5.2.5 The role of the βγ-dimer
- 5.2.6 Phosphorylation
- 5.3 Signal transduction involving G-proteins and phospholipase Cβ
- 5.3.1 G-protein effect on phospholipase Cβ
- 5.3.2 Action of the secondary messenger: diacylglycerol
- 5.3.3 Action of the secondary messenger: inositol triphosphate
- 5.3.4 Resynthesis of phosphatidylinositol diphosphate
- 5.4 The role of β-arrestins in modulating the activity of G-protein-coupled receptors
- 5.5 Signal transduction involving kinase receptors
- 5.5.1 Activation of signalling proteins and enzymes
- 5.5.2 The MAPK signal transduction pathway
- 5.5.3 Activation of guanylate cyclase by kinase receptors
- 5.5.4 The JAK-STAT signal transduction pathway
- 5.5.5 The PI3K/Akt/mTOR signal transduction pathway
- 5.6 The hedgehog signalling pathway
- Questions
- Further Reading
- List of Key Terms
- 6.1 Structure of DNA
- 6.1.1 The primary structure of DNA
- 6.1.2 The secondary structure of DNA
- 6.1.3 The tertiary structure of DNA
- 6.1.4 Chromatins
- 6.1.5 Genetic polymorphism and personalized medicine
- 6.2 Ribonucleic acid and protein synthesis
- 6.2.1 Structure of RNA
- 6.2.2 Transcription and translation
- 6.2.3 Small nuclear RNA
- 6.2.4 The regulatory role of RNA
- 6.3 Genetic illnesses
- 6.4 Molecular biology and genetic engineering
- Questions
- Further Reading
- List of Key Terms
- 7 Enzymes as drug targets
- 7.1 Inhibitors acting at the active site of an enzyme
- 7.1.1 Reversible inhibitors
- 7.1.2 Irreversible inhibitors
- 7.2 Inhibitors acting at allosteric binding sites
- 7.3 Uncompetitive and non-competitive inhibitors
- 7.4 Transition-state analogues: renin inhibitors
- 7.5 Suicide substrates
- 7.6 Isozyme selectivity of inhibitors
- 7.7 Medicinal uses of enzyme inhibitors
- 7.7.1 Enzyme inhibitors used against microorganisms
- 7.7.2 Enzyme inhibitors used against viruses
- 7.7.3 Enzyme inhibitors used against the body’s own enzymes
- 7.7.4 Enzyme modulators
- 7.8 Enzyme kinetics
- 7.8.1 Lineweaver–Burk plots
- 7.8.2 Comparison of inhibitors
- Questions
- Further Reading
- List of Key Terms
- 7.1 Inhibitors acting at the active site of an enzyme
- 8 Receptors as drug targets
- 8.1 Introduction
- 8.2 The design of agonists
- 8.2.1 Binding groups
- 8.2.2 Position of the binding groups
- 8.2.3 Size and shape
- 8.2.4 Other design strategies
- 8.2.5 Pharmacodynamics and pharmacokinetics
- 8.2.6 Examples of agonists
- 8.2.7 Allosteric modulators
- 8.3 The design of antagonists
- 8.3.1 Antagonists acting at the binding site
- 8.3.2 Antagonists acting outside the binding site
- 8.4 Partial agonists
- 8.5 Inverse agonists
- 8.6 Desensitization and sensitization
- 8.7 Tolerance and dependence
- 8.8 Receptor types and subtypes
- 8.9 Affinity, efficacy, and potency
- Questions
- Further Reading
- List of Key Terms
- 9 Nucleic acids as drug targets
- 9.1 Intercalating drugs acting on DNA
- 9.2 Topoisomerase poisons: non-intercalating
- 9.3 Alkylating and metallating agents
- 9.3.1 Nitrogen mustards
- 9.3.2 Nitrosoureas
- 9.3.3 Busulfan
- 9.3.4 Cisplatin
- 9.3.5 Dacarbazine and procarbazine
- 9.3.6 Mitomycin C
- 9.4 Chain cutters
- 9.5 Chain terminators
- 9.6 Control of gene transcription
- 9.7 Agents that act on RNA
- 9.7.1 Agents that bind to ribosomes
- 9.7.2 Antisense therapy
- Questions
- Further Reading
- List of Key Terms
- 10 Miscellaneous drug targets
- 10.1 Transport proteins as drug targets
- 10.2 Structural proteins as drug targets
- 10.2.1 Viral structural proteins as drug targets
- 10.2.2 Tubulin as a drug target
- 10.2.2.1 Agents that inhibit tubulin polymerization
- 10.2.2.2 Agents that inhibit tubulin depolymerization
- 10.3 Biosynthetic building blocks as drug targets
- 10.4 Biosynthetic processes as drug targets: chain terminators
- 10.5 Protein–protein interactions
- 10.5.1 Inhibition of protein–protein interactions
- 10.5.2 Promotion of protein–protein interactions
- 10.6 Lipids as a drug target
- 10.6.1 ‘Tunnelling molecules’
- 10.6.2 Ion carriers
- 10.6.3 Tethers and anchors
- 10.7 Carbohydrates as drug targets
- 10.7.1 Glycomics
- 10.7.2 Antigens and antibodies
- 10.7.3 Cyclodextrins
- Questions
- Further Reading
- List of Key Terms
- 11.1 The three phases of drug action
- 11.2 A typical journey for an orally active drug
- 11.3 Drug absorption
- 11.4 Drug distribution
- 11.4.1 Distribution round the blood supply
- 11.4.2 Distribution to tissues
- 11.4.3 Distribution to cells
- 11.4.4 Other distribution factors
- 11.4.5 Blood–brain barrier
- 11.4.6 Placental barrier
- 11.4.7 Drug–drug interactions
- 11.5 Drug metabolism
- 11.5.1 Phase I and phase II metabolism
- 11.5.2 Phase I transformations catalysed by cytochrome P450 enzymes
- 11.5.3 Phase I transformations catalysed by flavin-containing monooxygenases
- 11.5.4 Phase I transformations catalysed by other enzymes
- 11.5.5 Phase II transformations
- 11.5.6 Metabolic stability
- 11.5.7 The first pass effect
- 11.6 Drug excretion
- 11.7 Drug administration
- 11.7.1 Oral administration
- 11.7.2 Absorption through mucous membranes
- 11.7.3 Rectal administration
- 11.7.4 Topical administration
- 11.7.5 Inhalation
- 11.7.6 Injection
- 11.7.7 Implants
- 11.8 Drug dosing
- 11.8.1 Drug half-life
- 11.8.2 Steady state concentration
- 11.8.3 Drug tolerance
- 11.8.4 Bioavailability
- 11.9 Formulation
- 11.10 Drug delivery
- Questions
- Further Reading
- List of Key Terms
- CS1.1 Cholesterol and coronary heart disease
- CS1.2 The target enzyme
- CS1.3 The discovery of statins
- CS1.3.1 Type I statins
- CS1.3.2 Type II statins
- CS1.4 Mechanism of action for statins—pharmacodynamics
- CS1.5 Binding interactions of statins
- CS1.6 Other mechanisms of action for statins
- CS1.7 Other targets for cholesterol-lowering drugs
- Further Reading
- 12 Drug discovery: finding a lead
- 12.1 Choosing a disease
- 12.2 Choosing a drug target
- 12.2.1 Drug targets
- 12.2.2 Discovering drug targets
- 12.2.3 Target specificity and selectivity between species
- 12.2.4 Target specificity and selectivity within the body
- 12.2.5 Targeting drugs to specific organs and tissues
- 12.2.6 Pitfalls
- 12.2.7 Multi-target drugs
- 12.3 Identifying a bioassay
- 12.3.1 Choice of bioassay
- 12.3.2 In vitro tests
- 12.3.3 In vivo tests
- 12.3.4 Test validity
- 12.3.5 High-throughput screening
- 12.3.6 Screening by NMR
- 12.3.7 Affinity screening
- 12.3.8 Surface plasmon resonance
- 12.3.9 Scintillation proximity assay
- 12.3.10 Isothermal titration calorimetry
- 12.3.11 Virtual screening
- 12.4 Finding a lead compound
- 12.4.1 Screening of natural products
- 12.4.1.1 The plant kingdom
- 12.4.1.2 Microorganisms
- 12.4.1.3 Marine sources
- 12.4.1.4 Animal sources
- 12.4.1.5 Venoms and toxins
- 12.4.2 Medical folklore
- 12.4.3 Screening synthetic compound ‘libraries’
- 12.4.4 Existing drugs
- 12.4.4.1 ‘Me too’ and ‘me better’ drugs
- 12.4.4.2 Enhancing a side effect
- 12.4.5 Starting from the natural ligand or modulator
- 12.4.5.1 Natural ligands for receptors
- 12.4.5.2 Natural substrates for enzymes
- 12.4.5.3 Enzyme products as lead compounds
- 12.4.5.4 Natural modulators as lead compounds
- 12.4.6 Combinatorial and parallel synthesis
- 12.4.7 Computer-aided design of lead compounds
- 12.4.8 Serendipity and the prepared mind
- 12.4.9 Computerized searching of structural databases
- 12.4.10 Fragment-based lead discovery
- 12.4.11 Properties of lead compounds
- 12.4.1 Screening of natural products
- 12.5 Isolation and purification
- 12.6 Structure determination
- 12.7 Herbal medicine
- Questions
- Further Reading
- List of Key Terms
- 13 Drug design: optimizing target interactions
- 13.1 Structure–activity relationships
- 13.1.1 Binding role of alcohols and phenols
- 13.1.2 Binding role of aromatic rings
- 13.1.3 Binding role of alkenes
- 13.1.4 Binding role of ketones and aldehydes
- 13.1.5 Binding role of amines
- 13.1.6 Binding role of amides
- 13.1.7 Binding role of quaternary ammonium salts
- 13.1.8 Binding role of carboxylic acids
- 13.1.9 Binding role of esters
- 13.1.10 Binding role of alkyl and aryl halides
- 13.1.11 Binding role of thiols and ethers
- 13.1.12 Binding role of phosphates, phosphonates, and phosphinates
- 13.1.13 Binding role of other functional groups
- 13.1.14 Binding role of alkyl groups and the carbon skeleton
- 13.1.15 Binding role of heterocycles
- 13.1.16 Isosteres
- 13.1.17 Testing procedures
- 13.1.18 SAR in drug optimization
- 13.2 Identification of a pharmacophore
- 13.3 Drug optimization: strategies in drug design
- 13.3.1 Variation of substituents
- 13.3.1.1 Alkyl substituents
- 13.3.1.2 Substituents on aromatic or heteroaromatic rings
- 13.3.1.3 Varying substituents to change the pKa of ionizable groups
- 13.3.1.4 Synergistic effects
- 13.3.2 Extension of the structure
- 13.3.3 Chain extension/contraction
- 13.3.4 Ring expansion/contraction
- 13.3.5 Ring variations
- 13.3.6 Ring fusions
- 13.3.7 Isosteres and bio-isosteres
- 13.3.8 Simplification of the structure
- 13.3.9 Rigidification of the structure
- 13.3.10 Conformational blockers
- 13.3.11 Rigidification through intramolecular bonds
- 13.3.12 Structure-based drug design and molecular modelling
- 13.3.13 Drug design by NMR spectroscopy
- 13.3.14 The elements of luck and inspiration
- 13.3.15 Designing drugs to interact with more than one target
- 13.3.15.1 Agents designed from known drugs
- 13.3.15.2 Agents designed from non-selective lead compounds
- 13.3.1 Variation of substituents
- 13.1 Structure–activity relationships
- 13.4 Selectivity
- 13.5 Pharmacokinetics
- Questions
- Further Reading
- List of Key Terms
- 14.1 Optimizing hydrophilic/hydrophobic properties
- 14.1.1 Masking polar functional groups to decrease polarity
- 14.1.2 Adding or removing polar functional groups to vary polarity
- 14.1.3 Varying hydrophobic substituents to vary polarity
- 14.1.4 Variation of N-alkyl substituents to vary pKa
- 14.1.5 Other structural variations affecting pKa
- 14.1.6 Bio-isosteres for polar groups involved in binding interactions
- 14.2 Making drugs more resistant to chemical and enzymatic degradation
- 14.2.1 Steric shields
- 14.2.2 Electronic effects of bio-isosteres and substituents
- 14.2.3 Steric and electronic modifications
- 14.2.4 Metabolic blockers
- 14.2.5 Removal or replacement of susceptible metabolic groups
- 14.2.6 Group shifts
- 14.2.7 Ring variation and ring substituents
- 14.3 Making drugs less resistant to drug metabolism
- 14.3.1 Introducing metabolically susceptible groups
- 14.3.2 Self-destruct drugs
- 14.4 Targeting drugs
- 14.4.1 Targeting tumour cells: ‘search and destroy’ drugs
- 14.4.2 Targeting gastrointestinal infections
- 14.4.3 Targeting peripheral regions rather than the central nervous system
- 14.4.4 Targeting with membrane tethers
- 14.4.5 Targeting antibacterial agents using siderophores
- 14.5 Reducing toxicity
- 14.6 Prodrugs
- 14.6.1 Prodrugs to improve membrane permeability
- 14.6.1.1 Esters as prodrugs
- 14.6.1.2 N-Methylated prodrugs
- 14.6.1.3 Trojan horse approach for transport proteins
- 14.6.2 Prodrugs to prolong drug activity
- 14.6.3 Prodrugs masking drug toxicity and side effects
- 14.6.4 Prodrugs to lower water solubility
- 14.6.5 Prodrugs to improve water solubility
- 14.6.6 Prodrugs used in the targeting of drugs
- 14.6.7 Prodrugs to increase chemical stability
- 14.6.8 Prodrugs activated by external influence (sleeping agents)
- 14.6.1 Prodrugs to improve membrane permeability
- 14.7 Drug alliances
- 14.7.1 ‘Sentry’ drugs
- 14.7.2 Localizing a drug’s area of activity
- 14.7.3 Increasing absorption
- 14.8 Endogenous compounds as drugs
- 14.8.1 Neurotransmitters
- 14.8.2 Natural hormones, peptides, and proteins as drugs
- 14.9 Peptides and peptidomimetics in drug design
- 14.9.1 Peptidomimetics
- 14.9.2 Peptide drugs
- 14.10 Oligonucleotides as drugs
- Questions
- Further Reading
- List of Key Terms
- 15.1 Preclinical and clinical trials
- 15.1.1 Toxicity testing
- 15.1.2 Drug metabolism studies
- 15.1.3 Pharmacology, formulation, and stability tests
- 15.1.4 Clinical trials
- 15.1.4.1 Phase I studies
- 15.1.4.2 Phase II studies
- 15.1.4.3 Phase III studies
- 15.1.4.4 Phase IV studies
- 15.1.4.5 Ethical issues
- 15.2.1 Patents
- 15.2.2 Regulatory affairs
- 15.2.2.1 The regulatory process
- 15.2.2.2 Fast-tracking and orphan drugs
- 15.2.2.3 Good laboratory, manufacturing, and clinical practice
- 15.2.2.4 Cost-versus-benefit analysis
- 15.3.1 Chemical development
- 15.3.2 Process development
- 15.3.3 Choice of drug candidate
- 15.3.4 Natural products
- Further Reading
- CS3.1 Introduction
- CS3.2 Artemisinin
- CS3.3 Structure and synthesis of artemisinin
- CS3.4 Structure–activity relationships
- CS3.5 Mechanism of action
- CS3.6 Drug design and development
- Further Reading
- List of Key Terms
- CS4.1 Introduction
- CS4.2 From lucanthone to oxamniquine
- CS 4.3 Mechanism of action
- CS4.4 Other agents
- Further Reading
- CS5.1 Introduction
- CS5.2 The malarial parasite
- CS5.3 The target for fosmidomycin: DOXP reductoisomerase
- CS5.4 Fosmidomycin as a transition-state analogue
- CS5.5 Binding interactions of fosmidomycin
- CS5.6 Structure–activity relationships (SARs)
- CS5.7 Properties of fosmidomycin
- CS5.8 Analogues of fosmidomycin via an extension strategy
- CS5.9 Prodrugs of fosmidomycin
- Further Reading
- 16 Combinatorial and parallel synthesis
- 16.1 Combinatorial and parallel synthesis in medicinal chemistry projects
- 16.2 Solid-phase techniques
- 16.2.1 The solid support
- 16.2.2 The anchor/linker
- 16.2.3 Examples of solid-phase syntheses
- 16.3 Planning and designing a compound library
- 16.3.1 ‘Spider-like’ scaffolds
- 16.3.2 Designing ‘drug-like’ molecules
- 16.3.3 Synthesis of scaffolds
- 16.3.4 Substituent variation
- 16.3.5 Designing compound libraries for lead optimization
- 16.3.6 Computer-designed libraries
- 16.4 Testing for activity
- 16.4.1 High-throughput screening
- 16.4.2 Screening ‘on bead’ or ‘off bead’
- 16.5 Parallel synthesis
- 16.5.1 Solid-phase extraction
- 16.5.2 The use of resins in solution phase organic synthesis (SPOS)
- 16.5.3 Reagents attached to solid support: catch and release
- 16.5.4 Microwave technology
- 16.5.5 Microfluidics in parallel synthesis
- 16.6 Combinatorial synthesis
- 16.6.1 The mix and split method in combinatorial synthesis
- 16.6.2 Structure determination of the active compound(s)
- 16.6.2.1 Tagging
- 16.6.2.2 Photolithography
- 16.6.3 Dynamic combinatorial synthesis
- Questions
- Further Reading
- List of Key Terms
- 17 In silico drug design
- 17.1 Molecular and quantum mechanics
- 17.1.1 Molecular mechanics
- 17.1.2 Quantum mechanics
- 17.1.3 Choice of method
- 17.2 Drawing chemical structures
- 17.3 3D structures
- 17.4 Energy minimization
- 17.5 Viewing 3D molecules
- 17.6 Molecular dimensions
- 17.7 Molecular properties
- 17.7.1 Partial charges
- 17.7.2 Molecular electrostatic potentials
- 17.7.3 Molecular orbitals
- 17.7.4 Spectroscopic transitions
- 17.7.5 The use of grids in measuring molecular properties
- 17.8 Conformational analysis
- 17.8.1 Local and global energy minima
- 17.8.2 Molecular dynamics
- 17.8.3 Stepwise bond rotation
- 17.8.4 Monte Carlo and the Metropolis method
- 17.8.5 Genetic and evolutionary algorithms
- 17.9 Structure comparisons and overlays
- 17.10 Identifying the active conformation
- 17.10.1 X-ray crystallography
- 17.10.2 Comparison of rigid and non-rigid ligands
- 17.11 3D pharmacophore identification
- 17.11.1 X-ray crystallography
- 17.11.2 Structural comparison of active compounds
- 17.11.3 Automatic identification of pharmacophores
- 17.12 Docking procedures
- 17.12.1 Manual docking
- 17.12.2 Automatic docking
- 17.12.3 Defining the molecular surface of a binding site
- 17.12.4 Rigid docking by shape complementarity
- 17.12.5 The use of grids in docking programs
- 17.12.6 Rigid docking by matching hydrogen-bonding groups
- 17.12.7 Rigid docking of flexible ligands: the FLOG program
- 17.12.8 Docking of flexible ligands: anchor and grow programs
- 17.12.8.1 Directed Dock and Dock 4.0
- 17.12.8.2 FlexX
- 17.12.8.3 The Hammerhead program
- 17.12.9 Docking of flexible ligands: simulated annealing and genetic algorithms
- 17.13 Automated screening of databases for lead compounds and drug design
- 17.14 Protein mapping
- 17.14.1 Constructing a model protein: homology modelling
- 17.14.2 Constructing a binding site: hypothetical pseudoreceptors
- 17.15 De novo drug design
- 17.15.1 General principles of de novo drug design
- 17.15.2 Automated de novo drug design
- 17.15.2.1 LUDI
- Stage 1: Identification of interaction sites
- Stage 2: Fitting molecular fragments
- Stage 3: Fragment bridging
- 17.15.2.2 SPROUT
- 17.15.2.3 LEGEND
- 17.15.2.4 GROW, ALLEGROW, and SYNOPSIS
- 17.15.2.1 LUDI
- 17.1 Molecular and quantum mechanics
- 17.16 Planning compound libraries
- 17.17 Database handling
- Questions
- Further Reading
- List of Key Terms
- 18.1 Graphs and equations
- 18.2 Physicochemical properties
- 18.2.1 Hydrophobicity
- 18.2.1.1 The partition coefficient (P)
- 18.2.1.2 The substituent hydrophobicity constant (π)
- 18.2.1.3 P versus π
- 18.2.2 Electronic effects
- 18.2.3 Steric factors
- 18.2.3.1 Taft’s steric factor (Es)
- 18.2.3.2 Molar refractivity
- 18.2.3.3 Verloop steric parameter
- 18.2.4 Other physicochemical parameters
- 18.2.1 Hydrophobicity
- 18.3 Hansch equation
- 18.4 The Craig plot
- 18.5 The Topliss scheme
- 18.6 Bio-isosteres
- 18.7 The Free–Wilson approach
- 18.8 Planning a QSAR study
- 18.9 Case study: anti-allergic activity of a series of pyranenamines
- 18.10 3D QSAR
- 18.10.1 Defining steric and electrostatic fields
- 18.10.2 Relating shape and electronic distribution to biological activity
- 18.10.3 Advantages of CoMFA over traditional QSAR
- 18.10.4 Potential problems of CoMFA
- 18.10.5 Other 3D QSAR methods
- 18.10.6 Case study: inhibitors of tubulin polymerization
- Questions
- Further Reading
- List of Key Terms
- Further Reading
- List of Key Terms
- 19 Antibacterial agents
- 19.1 History of antibacterial agents
- 19.2 The bacterial cell
- 19.3 Mechanisms of antibacterial action
- 19.4 Antibacterial agents that act against cell metabolism (antimetabolites)
- 19.4.1 Sulphonamides
- 19.4.1.1 The history of sulphonamides
- 19.4.1.2 Structure–activity relationships
- 19.4.1.3 Sulphanilamide analogues
- 19.4.1.4 Applications of sulphonamides
- 19.4.1.5 Mechanism of action
- 19.4.2 Examples of other antimetabolites
- 19.4.2.1 Trimethoprim
- 19.4.2.2 Sulphones
- 19.4.1 Sulphonamides
- 19.5 Antibacterial agents that inhibit cell wall synthesis
- 19.5.1 Penicillins
- 19.5.1.1 History of penicillins
- 19.5.1.2 Structure of benzylpenicillin and phenoxymethylpenicillin
- 19.5.1.3 Properties of benzylpenicillin
- 19.5.1.4 Mechanism of action for penicillin
- Structure of the cell wall
- The transpeptidase enzyme and its inhibition
- 19.5.1.5 Resistance to penicillin
- Physical barriers
- Presence of β-lactamase enzymes
- High levels of transpeptidase enzyme produced
- Affinity of the transpeptidase enzyme to penicillin
- Transport back across the outer membrane of Gram-negative bacteria
- Mutations and genetic transfers
- 19.5.1.6 Methods of synthesizing penicillin analogues
- Fermentation
- Semi-synthetic procedure
- 19.5.1.7 Structure–activity relationships of penicillins
- 19.5.1.8 Penicillin analogues
- Acid sensitivity of penicillins
- Acid-resistant penicillins
- β-Lactamase-resistant penicillins
- Broad-spectrum penicillins
- Broad-spectrum penicillins: the aminopenicillins
- Broad-spectrum penicillins: the carboxypenicillins
- Broad-spectrum penicillins: the ureidopenicillins
- 19.5.1.9 Synergism of penicillins with other drugs
- 19.5.2 Cephalosporins
- 19.5.2.1 Cephalosporin C
- Discovery and structure of cephalosporin C
- Properties of cephalosporin C
- Structure–activity relationships of cephalosporin C
- 19.5.2.2 Synthesis of cephalosporin analogues at position 7
- 19.5.2.3 First-generation cephalosporins
- 19.5.2.4 Second-generation cephalosporins
- Cephamycins
- Oximinocephalosporins
- 19.5.2.5 Third-generation cephalosporins
- 19.5.2.6 Fourth-generation cephalosporins
- 19.5.2.7 Fifth-generation cephalosporins
- 19.5.2.8 Resistance to cephalosporins
- 19.5.2.1 Cephalosporin C
- 19.5.3 Other β-lactam antibiotics
- 19.5.3.1 Carbapenems
- 19.5.3.2 Monobactams
- 19.5.4 β-Lactamase inhibitors
- 19.5.4.1 Clavulanic acid
- 19.5.4.2 Penicillanic acid sulphone derivatives
- 19.5.4.3 Olivanic acids
- 19.5.4.4 β-Lactamase inhibitors lacking a β-lactam ring
- 19.5.5 Other drugs that act on bacterial cell wall biosynthesis
- 19.5.5.1 d-Cycloserine and bacitracin
- 19.5.5.2 The glycopeptides: vancomycin and vancomycin analogues
- 19.5.1 Penicillins
- 19.6.1 Valinomycin and gramicidin A
- 19.6.2 Polymyxin B
- 19.6.3 Killer nanotubes
- 19.6.4 Cyclic lipopeptides
- 19.7.1 Aminoglycosides
- 19.7.2 Tetracyclines
- 19.7.3 Chloramphenicol
- 19.7.4 Macrolides
- 19.7.5 Lincosamides
- 19.7.6 Streptogramins
- 19.7.7 Oxazolidinones
- 19.7.8 Pleuromutilins
- 19.8.1 Quinolones and fluoroquinolones
- 19.8.2 Spiropyrimidinetriones
- 19.8.3 Aminoacridines
- 19.8.4 Rifamycins
- 19.8.5 Nitroimidazoles and nitrofurantoin
- 19.8.6 Inhibitors of bacterial RNA polymerase
- 19.11.1 Drug resistance by mutation
- 19.11.2 Drug resistance by genetic transfer
- 19.11.3 Other factors affecting drug resistance
- 19.11.4 The way ahead
- 20.1 Viruses and viral diseases
- 20.2 Structure of viruses
- 20.3 Life cycle of viruses
- 20.4 Vaccination
- 20.5 Antiviral drugs: general principles
- 20.6 Antiviral drugs used against DNA viruses
- 20.6.1 Inhibitors of viral DNA polymerase
- 20.6.2 Inhibitors of the DNA terminase complex
- 20.6.3 Kinase inhibitors
- 20.6.4 Inhibitors of tubulin polymerization
- 20.6.5 Antisense therapy
- 20.6.6 Antiviral drugs acting against hepatitis B
- 20.7 Antiviral drugs acting against RNA viruses: the human immunodeficiency virus (HIV)
- 20.7.1 Structure and life cycle of HIV
- 20.7.2 Antiviral therapy against HIV
- 20.7.3 Inhibitors of viral reverse transcriptase
- 20.7.3.1 Nucleoside reverse transcriptase inhibitors
- 20.7.3.2 Non-nucleoside reverse transcriptase inhibitors
- 20.7.4 Protease inhibitors
- 20.7.4.1 The HIV protease enzyme
- 20.7.4.2 Design of HIV protease inhibitors
- 20.7.4.3 Saquinavir
- 20.7.4.4 Ritonavir and lopinavir
- 20.7.4.5 Indinavir
- 20.7.4.6 Nelfinavir
- 20.7.4.7 Palinavir
- 20.7.4.8 Amprenavir and darunavir
- 20.7.4.9 Atazanavir
- 20.7.4.10 Tipranavir
- 20.7.4.11 Alternative design strategies for antiviral drugs targeting the HIV protease enzyme
- 20.7.5 Integrase inhibitors
- 20.7.6 Cell entry inhibitors
- 20.7.6.1 Fusion inhibitors targeting the viral gp41 glycoprotein
- 20.7.6.2 Inhibitors of the viral glycoprotein gp120
- 20.7.6.3 Inhibitors of the host cell CD4 protein
- 20.7.6.4 Inhibitors of the host cell CCR5 chemokine receptor
- 20.8.1 Structure and life cycle of the influenza virus
- 20.8.2 Ion channel disrupters: adamantanes
- 20.8.3 Neuraminidase inhibitors
- 20.8.3.1 Structure and mechanism of neuraminidase
- 20.8.3.2 Transition-state inhibitors: development of zanamivir (Relenza™)
- 20.8.3.3 Transition-state inhibitors: 6-carboxamides
- 20.8.3.4 Carbocyclic analogues: development of oseltamivir (Tamiflu™)
- 20.8.3.5 Other ring systems
- 20.8.3.6 Resistance studies
- 20.8.4 Cap-dependent endonuclease inhibitors
- 20.10.1 Inhibitors of HCV NS3-4A protease
- 20.10.1.1 Introduction
- 20.10.1.2 Design of boceprevir and telaprevir
- 20.10.1.3 Second-generation protease inhibitors
- 20.10.2 Inhibitors of HCV NS5B RNA-dependent RNA polymerase
- 20.10.3 Inhibitors of HCV NS5A protein
- 20.10.3.1 Symmetrical inhibitors
- 20.10.3.2 Unsymmetrical inhibitors
- 20.10.4 Other targets
- 20.11.1 Agents acting against cytidine triphosphate synthetase
- 20.11.2 Agents acting against S-adenosylhomocysteine hydrolase
- 20.11.3 Ribavirin
- 20.11.4 Interferons
- 20.11.5 Antibodies and ribozymes
- 20.11.5.1 Antibodies
- 20.11.5.2 Ribozymes
- 21.1 Cancer: an introduction
- 21.1.1 Definitions
- 21.1.2 Causes of cancer
- 21.1.3 Genetic faults leading to cancer: proto-oncogenes and oncogenes
- 21.1.3.1 Activation of proto-oncogenes
- 21.1.3.2 Inactivation of tumour suppression genes (anti-oncogenes)
- 21.1.3.3 The consequences of genetic defects
- 21.1.4 Abnormal signalling pathways
- 21.1.5 Insensitivity to growth-inhibitory signals
- 21.1.6 Abnormalities in cell cycle regulation
- 21.1.7 Apoptosis and the p53 protein
- 21.1.8 Telomeres
- 21.1.9 Angiogenesis
- 21.1.10 Tissue invasion and metastasis
- 21.1.11 Treatment of cancer
- 21.1.12 Resistance
- 21.2 Drugs acting directly on nucleic acids
- 21.2.1 Intercalating agents
- 21.2.2 Non-intercalating agents that inhibit the action of topoisomerase enzymes on DNA
- 21.2.2.1 Podophyllotoxins
- 21.2.2.2 Camptothecins
- 21.2.3 Alkylating and metallating agents
- 21.2.3.1 Nitrogen mustards
- 21.2.3.2 Cisplatin and cisplatin analogues: metallating agents
- 21.2.3.3 CC 1065 analogues
- 21.2.3.4 Ecteinascidins
- 21.2.3.5 Other alkylating agents
- 21.2.4 Chain cutters
- 21.3 Drugs acting on enzymes: antimetabolites
- 21.3.1 Dihydrofolate reductase inhibitors
- 21.3.2 Inhibitors of thymidylate synthase
- 21.3.3 Inhibitors of ribonucleotide reductase
- 21.3.4 Inhibitors of adenosine deaminase
- 21.3.5 Cytidine deaminase inhibitors
- 21.3.6 Inhibitors of DNA polymerases
- 21.3.7 Purine antagonists
- 21.3.8 DNA methyltransferase inhibitors
- 21.4 Hormone-based therapies
- 21.4.1 Glucocorticoids, estrogens, progestins, and androgens
- 21.4.2 Luteinizing hormone-releasing hormone receptor agonists and antagonists
- 21.4.3 Anti-estrogens
- 21.4.4 Anti-androgens
- 21.4.5 Aromatase inhibitors
- 21.4.6 Mitotane
- 21.4.7 Somatostatin receptor agonists
- 21.5 Drugs acting on structural proteins
- 21.5.1 Agents that inhibit tubulin polymerization
- 21.5.2 Agents that inhibit tubulin depolymerization
- 21.6 Inhibitors of signalling pathways
- 21.6.1 Inhibition of farnesyl transferase and the Ras protein
- 21.6.1.1 Inhibition of farnesyl transferase
- 21.6.1.2 Inhibition of KRAS
- 21.6.2 Protein kinase inhibitors
- 21.6.3 Receptor antagonists of the hedgehog signalling pathway
- 21.6.1 Inhibition of farnesyl transferase and the Ras protein
- 21.7 Miscellaneous enzyme inhibitors
- 21.7.1 Matrix metalloproteinase inhibitors
- 21.7.2 Proteasome inhibitors
- 21.7.3 Histone deacetylase inhibitors
- 21.7.4 Inhibitors of poly ADP ribose polymerase
- 21.7.5 Isocitrate dehydrogenase inhibitors
- 21.7.6 Inhibitors for Enhancer of Zeste homologue 2
- 21.7.7 Other enzyme targets
- 21.8 Agents affecting pro-survival and pro-apoptotic proteins
- 21.9 Agents affecting nuclear transport proteins
- 21.10 Miscellaneous anticancer agents
- 21.10.1 Synthetic agents
- 21.10.2 Natural products
- 21.10.3 Modulation of transcription factor–coactivator interactions
- 21.10.4 PROTAC
- 21.11 Photodynamic therapy
- 21.12 Radionuclide therapy
- 21.13 The role of biologics in anticancer therapy
- Questions
- Further Reading
- List of Key Terms
- 22.1 Introduction
- 22.1.1 The active site of protein kinases
- 22.1.2 Classification of protein kinase inhibitors
- 22.1.3 Selectivity and resistance
- 22.2 Kinase inhibitors of the epidermal growth factor receptor (EGFR)
- 22.3 Kinase inhibitors of Abelson tyrosine kinase, c-KIT, PDGFR, and SRC
- 22.4 Kinase inhibitors of c-KIT and PDGFR
- 22.5 Inhibitors of cyclin-dependent kinases (CDKs)
- 22.6 Kinase inhibitors of the MAPK signal transduction pathway
- 22.6.1 BRAF inhibitors
- 22.6.2 MEK inhibitors
- 22.7 Kinase inhibitors of PI3K-PIP3 pathways
- 22.7.1 Introduction
- 22.7.2 Inhibitors of phosphoinositide 3-kinase
- 22.7.3 Inhibitors of Bruton’s tyrosine kinase
- 22.8 Kinase inhibitors of anaplastic lymphoma kinase (ALK)
- 22.9 Kinase inhibitors of RET and KIF5B-RET
- 22.10 Kinase inhibitors of Janus kinase
- 22.11 Kinase inhibitors of vascular endothelial growth factor receptor (VEGFR)
- 22.12 Kinase inhibitors of fibroblast growth factor receptor (FGFR)
- 22.13 Kinase inhibitors of neurotrophic tropomyosin receptor tyrosine kinase (NTRK or TRK) and ROS1
- 22.14 Kinase inhibitors of FMS-like tyrosine kinase 3 (FLT3)
- 22.15 Kinase inhibitors of colony-stimulating factor-1 receptor (CSF-1R)
- 22.16 Kinase inhibitors of the hepatocyte growth factor receptor (HGFR or c-MET)
- 22.17 Multi-receptor tyrosine kinase inhibitors
- 22.18 Kinase inhibition involving protein–protein binding interactions
- 22.19 Kinase degradation induced by protein–protein binding interactions
- 22.20 Summary
- Questions
- Further Reading
- List of Key Terms
- 23.1 The immune system
- 23.1.1 Introduction
- 23.1.2 Phagocytes
- 23.1.3 B-cells, plasma cells, and antibody production
- 23.1.4 T-cell lymphocytes
- 23.1.5 The complement system
- 23.1.6 Antibodies
- 23.2 Antibody manufacture
- 23.3 Monoclonal antibodies as anticancer agents
- 23.3.1 Introduction
- 23.3.2 Monoclonal antibodies targeting the HER family of receptors
- 23.3.3 Antibodies targeting VEGF and VEGFR
- 23.3.4 Antibodies targeting the RANK ligand
- 23.3.5 Antibodies targeting the glycolipid GD2
- 23.3.6 Antibodies targeting interleukins
- 23.3.7 Antibodies targeting CD proteins in B-cells
- 23.3.7.1 Antibodies targeting CD52
- 23.3.7.2 Antibodies targeting CD20
- 23.3.7.3 Antibodies targeting CD19
- 23.3.8 Antibodies targeting CD proteins on plasma cells
- 23.3.8.1 Antibodies targeting SLAMF7 (CD319)
- 23.3.8.2 Antibodies targeting CD38
- 23.3.9 Antibodies targeting the CCR4 receptor
- 23.3.10 Monoclonal antibodies acting on immune checkpoints
- 23.3.10.1 Introduction
- 23.3.10.2 Antibodies targeting CTLA-4
- 23.3.10.3 Antibodies targeting the programmed death receptor (PD-1)
- 23.3.10.4 Antibodies targeting programmed death ligand 1
- 23.4.1 Introduction
- 23.4.2 Antibody–drug conjugates targeting HER2
- 23.4.3 Antibody–drug conjugates targeting trophoblast cell-surface antigen
- 23.4.4 Antibody–drug conjugates targeting nectin 4
- 23.4.5 Antibody–drug conjugates targeting B-cells
- 23.4.5.1 Antibodies targeting CD19
- 23.4.5.2 Antibodies targeting CD20
- 23.4.5.3 Antibodies targeting CD22
- 23.4.5.4 Antibodies targeting CD30
- 23.4.5.5 Antibodies targeting CD33
- 23.4.5.6 Antibodies targeting CD79
- 23.4.5.7 Antibodies targeting CD142
- 23.8.1 Gene therapy
- 23.8.2 Antisense therapy
- 24.1 The peripheral nervous system
- 24.2 Motor nerves of the peripheral nervous system
- 24.2.1 The somatic motor nervous system
- 24.2.2 The autonomic motor nervous system
- 24.2.3 The enteric system
- 24.2.4 Defects in motor nerve transmission
- 24.3 The cholinergic system
- 24.3.1 The cholinergic signalling system
- 24.3.2 Presynaptic control systems
- 24.3.3 Co-transmitters
- 24.4 Agonists at the cholinergic receptor
- 24.5 Acetylcholine: structure, SAR, and receptor binding
- 24.6 The instability of acetylcholine
- 24.7 Design of acetylcholine analogues
- 24.7.1 Steric shields
- 24.7.2 Electronic effects
- 24.7.3 Combining steric and electronic effects
- 24.8 Clinical uses for cholinergic agonists
- 24.8.1 Muscarinic agonists
- 24.8.2 Nicotinic agonists
- 24.9 Antagonists of the muscarinic cholinergic receptor
- 24.9.1 Actions and uses of muscarinic antagonists
- 24.9.2 Muscarinic antagonists
- 24.9.2.1 Atropine and hyoscine
- 24.9.2.2 Structural analogues of atropine and hyoscine
- 24.9.2.3 Simplified analogues of atropine
- 24.9.2.4 Quinuclidine muscarinic agents
- 24.9.2.5 Other muscarinic antagonists
- 24.10.1 Applications of nicotinic antagonists
- 24.10.2 Nicotinic antagonists
- 24.10.2.1 Curare and tubocurarine
- 24.10.2.2 Decamethonium and suxamethonium
- 24.10.2.3 Steroidal neuromuscular blocking agents
- 24.10.2.4 Atracurium and mivacurium
- 24.10.2.5 Other nicotinic antagonists
- 24.12.1 Effect of anticholinesterases
- 24.12.2 Structure of the acetylcholinesterase enzyme
- 24.12.3 The active site of acetylcholinesterase
- 24.12.3.1 Crucial amino acids within the active site
- 24.12.3.2 Mechanism of hydrolysis
- 24.13.1 Carbamates
- 24.13.1.1 Physostigmine
- 24.13.1.2 Analogues of physostigmine
- 24.13.2 Organophosphorus compounds
- 24.13.2.1 Nerve agents
- 24.13.2.2 Medicines
- 24.13.2.3 Insecticides
- 24.15.1 Acetylcholinesterase inhibitors
- 24.15.2 Dual-action agents acting on the acetylcholinesterase enzyme
- 24.15.3 Multi-targeted agents acting on the acetylcholinesterase enzyme and the muscarinic M2 receptor
- 25.1 The adrenergic nervous system
- 25.1.1 Peripheral nervous system
- 25.1.2 Central nervous system
- 25.2 Adrenergic receptors
- 25.2.1 Types of adrenergic receptor
- 25.2.2 Distribution of receptors
- 25.3 Endogenous agonists for the adrenergic receptors
- 25.4 Biosynthesis of catecholamines
- 25.5 Metabolism of catecholamines
- 25.6 Neurotransmission
- 25.6.1 The neurotransmission process
- 25.6.2 Co-transmitters
- 25.6.3 Presynaptic receptors and control
- 25.7 Drug targets
- 25.8 The adrenergic binding site
- 25.9 Structure–activity relationships (SARs)
- 25.9.1 Important binding groups on catecholamines
- 25.9.2 Selectivity for α- versus β-adrenoceptors
- 25.10 Adrenergic agonists
- 25.10.1 General adrenergic agonists
- 25.10.2 α1-, α2-, β1-, and β3-agonists
- 25.10.3 β2-agonists and the treatment of asthma
- 25.11 Adrenergic receptor antagonists
- 25.11.1 General α/β-blockers
- 25.11.2 α-blockers
- 25.11.3 β-blockers as cardiovascular drugs
- 25.11.3.1 First-generation β-blockers
- 25.11.3.2 Structure–activity relationships of aryloxypropanolamines
- 25.11.3.3 Selective β1-blockers (second-generation β-blockers)
- 25.11.3.4 Short-acting β-blockers
- 25.12.1 Drugs that affect the biosynthesis of adrenergics
- 25.12.2 Drugs inhibiting the uptake of noradrenaline into storage vesicles
- 25.12.3 Release of noradrenaline from storage vesicles
- 25.12.4 Reuptake inhibitors of noradrenaline into presynaptic neurons
- 25.12.5 Inhibition of metabolic enzymes
- 26.1 History of opium
- 26.2 The active principle: morphine
- 26.2.1 Isolation of morphine
- 26.2.2 Structure and properties
- 26.3 Structure–activity relationships
- 26.4 The molecular target for morphine: opioid receptors
- 26.5 Morphine: pharmacodynamics and pharmacokinetics
- 26.6 Morphine analogues
- 26.6.1 Variation of substituents
- 26.6.2 Drug extension
- 26.6.3 Simplification or drug dissection
- 26.6.3.1 Removing ring E
- 26.6.3.2 Removing ring D
- 26.6.3.3 Removing rings C and D
- 26.6.3.4 Removing rings B, C, and D
- 26.6.3.5 Removing rings B, C, D, and E
- 26.6.4 Rigidification
- 26.7 Agonists and antagonists
- 26.8 Endogenous opioid peptides and opioids
- 26.8.1 Endogenous opioid peptides
- 26.8.2 Analogues of enkephalins and δ-selective opioids
- 26.8.3 Binding theories for enkephalins
- 26.8.4 Inhibitors of peptidases
- 26.8.5 Endogenous morphine
- 26.9 The future
- 26.9.1 The message–address concept
- 26.9.2 Receptor dimers
- 26.9.3 Selective opioid agonists versus multi-targeted opioids
- 26.9.4 Peripheral-acting opioids
- 26.9.5 Biased agonists
- 26.10 Case study: design of nalfurafine
- Questions
- Further Reading
- List of Key Terms
- 27.1 Peptic ulcers
- 27.1.1 Definition
- 27.1.2 Causes
- 27.1.3 Treatment
- 27.1.4 Gastric acid release
- 27.2 H2-Antagonists
- 27.2.1 Histamine and histamine receptors
- 27.2.2 Searching for a lead
- 27.2.2.1 Histamine
- 27.2.2.2 Nα-Guanylhistamine
- 27.2.3 Developing the lead: a chelation bonding theory
- 27.2.4 From partial agonist to antagonist: the development of burimamide
- 27.2.5 Development of metiamide
- 27.2.6 Development of cimetidine
- 27.2.7 Cimetidine
- 27.2.7.1 Biological activity
- 27.2.7.2 Structure and activity
- 27.2.7.3 Metabolism
- 27.2.8 Further studies of cimetidine analogues
- 27.2.8.1 Conformational isomers
- 27.2.8.2 Desolvation
- 27.2.8.3 Development of the nitroketeneaminal binding group
- 27.2.9 Further H2-antagonists
- 27.2.9.1 Ranitidine
- 27.2.9.2 Famotidine and nizatidine
- 27.2.9.3 H2-Antagonists with prolonged activity
- 27.2.10 Comparison of H1- and H2-antagonists
- 27.2.11 H2 Receptors and H2-antagonists
- 27.3 Proton pump inhibitors (PPIs)
- 27.3.1 Parietal cells and the proton pump
- 27.3.2 PPIs
- 27.3.3 Mechanism of inhibition
- 27.3.4 Metabolism of PPIs
- 27.3.5 Design of omeprazole and esomeprazole
- 27.3.6 Other PPIs
- 27.4 Helicobacter pylori and the use of antibacterial agents
- 27.4.1 Discovery of Helicobacter pylori
- 27.4.2 Treatment
- 27.5 Traditional and herbal medicines
- Questions
- Further Reading
- List of Key Terms
- 28.1 Introduction
- 28.2 The cardiovascular system
- 28.3 Antihypertensives affecting the activity of the RAAS system
- 28.3.1 Introduction
- 28.3.2 Renin inhibitors
- 28.3.3 ACE inhibitors
- 28.3.4 Angiotensin receptor antagonists
- 28.3.5 Mineralocorticoid receptor antagonists
- 28.3.6 Dual-action agents
- 28.4 Endothelin receptor antagonists as antihypertensive agents
- 28.4.1 Endothelins and endothelin receptors
- 28.4.2 Endothelin antagonists
- 28.4.3 Dual-action agents
- 28.5 Vasodilators
- 28.5.1 Modulators of soluble guanylate cyclase
- 28.5.2 Phosphodiesterase type 5 inhibitors
- 28.5.3 Neprilysin inhibitors
- 28.5.4 Prostacyclin agonists
- 28.5.5 Miscellaneous vasodilators
- 28.6 Calcium entry blockers
- 28.6.1 Introduction
- 28.6.2 Dihydropyridines
- 28.6.3 Phenylalkylamines
- 28.6.4 Benzothiazepines
- 28.7 Funny ion channel inhibitors
- 28.8 Lipid-regulating agents
- 28.8.1 Statins
- 28.8.2 Fibrates
- 28.8.3 Dual- and pan-PPAR agonists
- 28.8.4 Antisense drugs
- 28.8.5 Inhibitors of transfer proteins
- 28.8.6 Antibodies as lipid-lowering agents
- 28.8.7 ATP citrate lyase inhibitors
- 28.9 Antithrombotic agents
- 28.9.1 Anticoagulants
- 28.9.1.1 Introduction
- 28.9.1.2 Direct thrombin inhibitors
- 28.9.1.3 Factor Xa inhibitors
- 28.9.2 Antiplatelet agents
- 28.9.2.1 Introduction
- 28.9.2.2 PAR-1 antagonists
- 28.9.2.3 P2Y12-Antagonists
- 28.9.2.4 GpIIb/IIIa antagonists
- 28.9.3 Fibrinolytic drugs
- 28.9.1 Anticoagulants
- Questions
- Further Reading
- List of Key Terms
- CS7.1 Introduction to steroids
- CS7.2 Orally active analogues of cortisol
- CS7.3 Topical glucocorticoids as anti-inflammatory agents
- CS7.3.1 Cortisol analogues
- CS7.3.2 21-Deoxysteroids
- CS7.3.3 11-Ketosteroids
- CS7.3.4 Analogues with modified C-17 side chains
- CS7.3.5 Glucocorticoids used in asthma treatment
- CS7.3.6 Glucocorticoids used in ophthalmology
- CS7.3.7 Sustained release of topical anti-inflammatory agents
- List of Key Terms
- CS8.1 Introduction
- CS8.2 The monoamine hypothesis
- CS8.3 Current antidepressant agents
- CS8.4 Current areas of research
- CS8.5 Antagonists for the 5-HT7 receptor
- Further Reading
- List of Key Terms
- CS9.1 Introduction
- CS9.2 Reaction catalysed by renin
- CS9.3 From lead compound to peptide inhibitors
- CS9.4 Peptidomimetic strategies
- CS9.5 Design of non-peptide inhibitors
- CS9.6 Optimization of the structure
- Further Reading
- List of Key Terms
- CS10.1 Introduction
- CS10.2 The target
- CS10.3 General strategies in the design of factor Xa inhibitors
- CS10.4 Apixaban: from hit structure to lead compound
- CS10.5 Apixaban: from lead compound to final structure
- CS10.6 The development of rivaroxaban
- CS10.7 The development of edoxaban
- Further Reading
- CS11.1 Introduction
- CS11.2 Identification of a lead compound
- CS11.3 Modifications of the lead compound
- CS11.4 From hexapeptide to tripeptide
- CS11.5 From tripeptide to macrocycle (BILN-2061)
- CS11.6 From BILN-2061 to simeprevir
- Further Reading
- List of Key Terms
- 1 Drugs and drug targets: an overview
- 2 Protein structure and function
- 3 Enzymes: structure and function
- 4 Receptors: structure and function
- 5 Receptors and signal transduction
- 6 Nucleic acids: structure and function
- 7 Enzymes as drug targets
- 8 Receptors as drug targets
- 9 Nucleic acids as drug targets
- 10 Miscellaneous drug targets
- 11 Pharmacokinetics and related topics
- Case study 1 Statins
- 12 Drug discovery: finding a lead
- 13 Drug design: optimizing target interactions
- 14 Drug design: optimizing access to the target
- 15 Getting the drug to market
- Case study 2 The design of ACE inhibitors
- Case study 3 Artemisinin and related antimalarial drugs
- Case study 4 The design of oxamniquine
- Case study 5 Fosmidomycin as an antimalarial agent
- 16 Combinatorial and parallel synthesis
- 17 In silico drug design
- 18 Quantitative structure–activity relationships (QSAR)
- Case study 6 Design of a thymidylate synthase inhibitor
- 19 Antibacterial agents
- 20 Antiviral agents
- 21 Anticancer agents
- 22 Protein kinase inhibitors as anticancer agents
- 23 Antibodies and other biologics
- 24 Cholinergics, anticholinergics, and anticholinesterases
- 25 Drugs acting on the adrenergic nervous system
- 26 The opioid analgesics
- 27 Anti-ulcer agents
- 28 Cardiovascular drugs
- Case Study 7 Steroidal anti-inflammatory agents
- Case Study 8 Current research into antidepressant agents
- Case Study 9 The design and development of aliskiren
- Case Study 10 Factor Xa inhibitors
- Case Study 11 Reversible inhibitors of HCV NS3-4A protease
- Appendix 1
UM RAFBÆKUR Á HEIMKAUP.IS
Bókahillan þín er þitt svæði og þar eru bækurnar þínar geymdar. Þú kemst í bókahilluna þína hvar og hvenær sem er í tölvu eða snjalltæki. Einfalt og þægilegt!Rafbók til eignar
Rafbók til eignar þarf að hlaða niður á þau tæki sem þú vilt nota innan eins árs frá því bókin er keypt.
Þú kemst í bækurnar hvar sem er
Þú getur nálgast allar raf(skóla)bækurnar þínar á einu augabragði, hvar og hvenær sem er í bókahillunni þinni. Engin taska, enginn kyndill og ekkert vesen (hvað þá yfirvigt).
Auðvelt að fletta og leita
Þú getur flakkað milli síðna og kafla eins og þér hentar best og farið beint í ákveðna kafla úr efnisyfirlitinu. Í leitinni finnur þú orð, kafla eða síður í einum smelli.
Glósur og yfirstrikanir
Þú getur auðkennt textabrot með mismunandi litum og skrifað glósur að vild í rafbókina. Þú getur jafnvel séð glósur og yfirstrikanir hjá bekkjarsystkinum og kennara ef þeir leyfa það. Allt á einum stað.
Hvað viltu sjá? / Þú ræður hvernig síðan lítur út
Þú lagar síðuna að þínum þörfum. Stækkaðu eða minnkaðu myndir og texta með multi-level zoom til að sjá síðuna eins og þér hentar best í þínu námi.
Fleiri góðir kostir
- Þú getur prentað síður úr bókinni (innan þeirra marka sem útgefandinn setur)
- Möguleiki á tengingu við annað stafrænt og gagnvirkt efni, svo sem myndbönd eða spurningar úr efninu
- Auðvelt að afrita og líma efni/texta fyrir t.d. heimaverkefni eða ritgerðir
- Styður tækni sem hjálpar nemendum með sjón- eða heyrnarskerðingu
- Gerð : 208
- Höfundur : 6450
- Útgáfuár : 2017
- Leyfi : 380