Cover image for Computational and structural approaches to drug discovery : ligand-protein interactions
Title:
Computational and structural approaches to drug discovery : ligand-protein interactions
Series:
RSC biomolecular sciences
Publication Information:
Cambridge : RSC Publishing, 2008
Physical Description:
xvii, 382 p. : ill. ; 25 cm.
ISBN:
9780854043651

Available:*

Library
Item Barcode
Call Number
Material Type
Item Category 1
Status
Searching...
30000010167849 RM301.42 C65 2008 Open Access Book Book
Searching...

On Order

Summary

Summary

Computational methods impact all aspects of modern drug discovery and most notably these methods move rapidly from academic exercises to becoming drugs in clinical trials... This insightful book represents the experience and understanding of the global experts in the field and spotlights both the structural and medicinal chemistry aspects of drug design. The need to 'encode' the factors that determine adsorption, distribution, metabolism, excretion and toxicology are explored, as they remain the critical issues in this area of research. This indispensable resource provides the reader with: * A rich understanding of modern approaches to docking * A comparison and critical evaluation of state-of-the-art methods * Details on harnessing computational methods for both analysis and prediction * An insight into prediction potencies and protocols for unbiased evaluations of docking and scoring algorithms * Critical reviews of current fragment based methods with perceptive applications to kinases Addressing a wide range of uses of protein structures for drug discovery the Editors have created and essential reference for professionals in the pharmaceutical industry and moreover an indispensable core text for all graduate level courses covering molecular interactions and drug discovery.


Author Notes

Robert M. Stroud is a professor at the University of California and has been a fellow of the Royal Society of Medicine (UK) since 1992 and a member of the National Academt of Sciences (US) since 2003. His prestigious career spans over 35 years and he is and has served on the scientific advisory boards of many companies and institutions including the National Cancer Institute, the Neutron Diffraction facility, Axys Pharmaceuticals, and Sunesis Phamraceuticals. Janet Finer-Moore is a Research Biologist at the University of California Her contribution to the detailed determination of the structural and chemical mechanism of a two substrate enzyme and detection of amphipathic helices in protein and gene sequences have perpetuated over 28 publications. She is a member of the AAAS, the ACA, the ACS and the Biophysical Society.


Table of Contents

Section 1 Overview
Chapter 1 Facing the Wall in Computationally Based Approaches to Drug DiscoveryJanet S. Finer-Moore and Jeff Blaney and Robert M. Stroud
1.1 The Promise, and the Problemp. 3
1.2 Current Limitations in Structure-guided Lead Designp. 5
1.3 Lessons in Structure-based Drug Design from Thymidylate Synthasep. 7
1.3.1 Mechanism-based Inhibitors and Enzyme-catalyzed Therapeuticsp. 7
1.3.2 Iterative Structure-based Drug Designp. 8
1.3.3 Docking, Fragments and Optimizabilityp. 8
1.4 New Developments in Structure-based Drug-design Methodsp. 13
1.4.1 Fragment-based Methodsp. 13
1.4.2 Identifying Drug Target Sites on a Proteinp. 16
1.4.3 Targeting Protein-Protein Interactionsp. 17
1.4.4 Computational Docking to Nominated Sitesp. 18
1.5 Conclusionp. 19
Referencesp. 20
Chapter 2 The Changing Landscape in Drug DiscoveryHugo Kubinyi
2.1 Introductionp. 24
2.2 QSAR - Understanding Without Predictionp. 25
2.3 Gene Technology - from Mice to Humansp. 27
2.4 Combinatorial Library Design - Driven by Medicinal Chemistryp. 28
2.5 Docking and Scoring - Solved and Unsolved Problemsp. 32
2.6 Virtual Screening - the Road to Successp. 35
2.7 Fragment-based and Combinatorial Design - A New Challengep. 37
2.8 Summary and Conclusionsp. 38
Referencesp. 41
Section 2 Structure-Based Design
Chapter 3 Purine Nucleoside PhosphorylaseYang Zhang and Steven E. Ealick
3.1 Introductionp. 49
3.2 Three-dimensional Structures of PNPsp. 51
3.3 Related Enzymes of the PNP Familyp. 54
3.4 PNP Active Sitesp. 55
3.5 Human PNP Inhibitorsp. 58
3.6 Other Applications of Molecular Design to PNPp. 62
3.7 Applications of Molecular Design to Enzymes Related to PNPp. 64
3.8 PNP Inhibitors and Clinical Trialsp. 65
3.9 Conclusions and Future Directionsp. 66
Note Added in Proofp. 66
Referencesp. 67
Chapter 4 Application and Limitations of X-Ray Crystallographic Data in Structure-Guided Ligand and Drug DesignAndrew M. Davis and Simon J. Teague and Gerard J. Kleywegt
4.1 Introductionp. 73
4.2 Structure-guided Ligand Design and Drug Designp. 74
4.3 Some Limitations in the Use of X-ray Datap. 79
4.3.1 Basic Crystallography Termsp. 79
4.3.2 Uncertainty in the Identity or Location of Protein or Ligand Atomsp. 83
4.3.3 Effect of Crystallization Conditionsp. 86
4.3.4 Identification and Location of Waterp. 87
4.4 Macromolecular Structures to Determine Small-molecule Structuresp. 88
4.5 Assessing the Validity of Structure Modelsp. 89
4.6 Summary and Outlookp. 90
Referencesp. 91
Chapter 5 Dealing with Bound Waters in a Site: Do they Leave or Stay?Donald Hamelberg and J. Andrew McCammon
5.1 Introductionp. 95
5.2 Localized Water Molecules in Binding Sites of Proteinsp. 96
5.3 Identifying Localized Water Molecules from Computer Simulationsp. 99
5.4 Calculation of Free-energy Cost of Displacing a Site-bound Water Moleculep. 101
5.5 Inclusion of Explicit Water Molecules in Drug Discoveryp. 104
Acknowledgementsp. 106
Referencesp. 106
Chapter 6 Knowledge-Based Methods in Structure-Based DesignMarcel L. Verdonk and Wijnand T.M. Mooij
6.1 Introductionp. 111
6.2 Atom-based Potentialsp. 111
6.3 Group-based Potentialsp. 112
6.4 Methodologiesp. 114
6.4.1 The Reference Statep. 115
6.4.2 Volume Correctionsp. 116
6.5 Applicationsp. 117
6.5.1 Visualization and Interaction 'Hot Spots'p. 117
6.5.2 Docking and Scoringp. 118
6.5.3 De Novo Designp. 120
6.5.4 Targeted Scoring Functionsp. 120
6.6 Discussionp. 121
6.7 Conclusionp. 123
Referencesp. 123
Chapter 7 Combating Drug Resistance - Identifying Resilient Molecular Targets and Robust DrugsCelia A. Schiffer
7.1 Introductionp. 127
7.2 Resilient Targets and Robust Drugsp. 128
7.3 Example of HIV-1 Protease: Substrate Recognition vs. Drug Resistancep. 129
7.4 Implications for Future Structure-based Drug Designp. 132
Acknowledgementsp. 132
Referencesp. 132
Section 3 Docking
Chapter 8 Docking Algorithms and Scoring Functions; State-of-the-Art and Current LimitationsGregory L. Warren and Catherine E. Peishoff and Martha S. Head
8.1 Introductionp. 137
8.1.1 Binding Mode Predictionp. 138
8.1.2 Virtual Screening for Lead Identificationp. 139
8.1.3 Potency Prediction for Lead Optimizationp. 139
8.2 A Brief Review of Recent Docking Evaluationsp. 140
8.3 What these Evaluations Tell us about the Performance of Docking Algorithmsp. 143
8.3.1 Binding Mode Predictionsp. 143
8.3.2 Virtual Screeningp. 144
8.3.3 Affinity Predictionp. 145
8.4 How an Ideal Evaluation Data Set Might be Structuredp. 147
8.4.1 Binding Mode Predictionp. 147
8.4.2 Virtual Screeningp. 148
8.4.3 Affinity Predictionp. 148
8.5 Concluding Remarksp. 149
8.5.1 Binding Mode Predictionp. 149
8.5.2 Virtual Screeningp. 150
8.5.3 Rank Order by Affinityp. 151
8.5.4 The State-of-the-artp. 152
Referencesp. 153
Chapter 9 Application of Docking Methods to Structure-Based Drug DesignDemetri T. Moustakas
9.1 Introductionp. 155
9.2 Docking Methods, Capabilities and Limitationsp. 156
9.2.1 Molecule Preparationp. 156
9.2.2 Sampling Methodsp. 157
9.2.3 Scoring Methodsp. 160
9.2.4 Managing Errors in Dockingp. 162
9.3 How is Docking Applied to Drug Design?p. 164
9.3.1 Drug Target Selection and Characterizationp. 165
9.3.2 Lead Compound Discoveryp. 168
9.3.3 Lead Compound Optimizationp. 171
9.4 Summaryp. 172
Referencesp. 172
Chapter 10 Strength in Flexibility: Modeling Side-Chain Conformational Change in Docking and ScreeningLeslie A. Kuhn
10.1 Introductionp. 181
10.2 Backgroundp. 181
10.2.1 Improving Docking and Screening Through Side-chain Flexibility Modelingp. 181
10.2.2 Enhancing Target Specificity Through Flexibility Modelingp. 182
10.3 Approachesp. 183
10.3.1 The State of the Art in Modeling Protein Side-chain Flexibilityp. 183
10.3.2 Learning from Nature: Observing Side-chain Motions Upon Ligand Bindingp. 185
10.4 The Future: Knowledge-based Modeling of Side-chain Motionsp. 189
Acknowledgementsp. 189
Referencesp. 190
Chapter 11 Avoiding the Rigid Receptor: Side-Chain RotamersAmy C. Anderson
11.1 Introductionp. 192
11.2 Rotamer Librariesp. 194
11.3 Successful Applications of Rotamer Libraries in Drug Designp. 195
11.3.1 Aspartic Acid Protease Inhibitorsp. 195
11.3.2 Matrix Metalloproteinase-1 Inhibitorsp. 195
11.3.3 Thymidylate Synthase Inhibitorsp. 199
11.3.4 Protein Tyrosine Phosphatase 1B Inhibitorsp. 200
11.3.5 HIV Protease Drug-resistant Mutants Bound to Inhibitorsp. 201
11.3.6 Trypsin-benzamidine and Phosphocholine-McPC 603p. 201
11.4 Conclusionsp. 202
Acknowledgementsp. 202
Referencesp. 202
Section 4 Screening
Chapter 12 Computational Prediction of Aqueous Solubility, Oral Bioavailability, P450 Activity and hERG Channel BlockadeDavid E. Clark
12.1 Introductionp. 207
12.2 Aqueous Solubilityp. 208
12.3 Oral Bioavailabilityp. 211
12.4 Cytochrome P450 Activityp. 212
12.5 hERG Channel Blockadep. 215
12.6 Conclusionsp. 219
Referencesp. 220
Chapter 13 Shadows on ScreensBrian K. Shoichet and Brian Y. Feng and Kristin E.D. Coan
13.1 Introductionp. 223
13.2 Phenomenology of Aggregationp. 224
13.3 What Sort of Compounds Aggregate?p. 227
13.4 Mechanism of Aggregation-based Inhibitionp. 232
13.5 A Rapid Counter-screen for Aggregation-based Inhibitorsp. 233
13.6 Biological Implications?p. 239
13.7 The Spirit-haunted World of Screeningp. 239
Acknowledgementsp. 240
Referencesp. 240
Chapter 14 Iterative Docking Strategies for Virtual Ligand ScreeningAlbert E. Beuscher IV and Arthur J. Olson
14.1 Introductionp. 242
14.2 AutoDock Backgroundp. 243
14.2.1 Scoring Functionp. 243
14.2.2 Search Functionp. 244
14.2.3 AutoDockToolsp. 244
14.2.4 AutoDockTools Analysisp. 245
14.3 Diversity-based Virtual Screening Studiesp. 246
14.3.1 AICAR Transformylasep. 246
14.3.2 Protein Phosphatase 2Cp. 246
14.4 Comparison with Existing VLS Strategiesp. 253
14.4.1 Hierarchical VLSp. 256
14.4.2 Monolithic VLS Strategyp. 258
14.5 Other AutoDock VLS Studiesp. 259
14.5.1 Acetylcholine Esterase Peripheral Anionic Sitep. 259
14.5.2 Human P2Y[subscript 1] Receptorp. 260
14.6 Diversity-based vs. Issuesp. 260
14.6.1 Library Choicep. 260
14.6.2 Similarity Searchp. 261
14.6.3 Apo Versus Ligand-bound Docking Modelsp. 262
14.6.4 Binding Site Choicesp. 263
14.7 Future Workp. 264
Referencesp. 264
Chapter 15 Challenges and Progresses in Calculations of Binding Free Energies - What Does it Take to Quantify Electrostatic Contributions to Protein-Ligand Interactions?Mitsunori Kato and Sonja Braun-Sand and Arieh Warshel
15.1 Introductionp. 268
15.2 Computational Strategiesp. 269
15.2.1 Free-energy Perturbation, Linear Response Approximation and Potential of Mean Force Calculations by All-atom Modelsp. 269
15.2.2 Proper and Improper Treatments of Long-range Effects in All-atom Modelsp. 273
15.2.3 Calculations of Electrostatic Energies by Simplified Modelsp. 274
15.3 Calculating Binding Free Energiesp. 277
15.3.1 Studies of Drug Mutations by FEP Approachesp. 277
15.3.2 Evaluation of Absolute Binding Energies by the LRA and LIE Approachesp. 278
15.3.3 Using Semi-macroscopic and Macroscopic Approaches in Studies of Ligand Bindingp. 279
15.3.4 Protein-protein Interactionsp. 281
15.4 Challenges and New Advancesp. 282
15.5 Perspectivesp. 285
Acknowledgementp. 285
Referencesp. 285
Section 5 Fragment-Based Design
Chapter 16 Discovery and Extrapolation of Fragment Structures towards Drug DesignAlessio Ciulli and Tom L. Blundell and Chris Abell
16.1 Structure-based Approaches to Drug Discoveryp. 293
16.2 Properties of Molecular Fragmentsp. 294
16.3 From Molecular Fragments to Drug Leadsp. 296
16.3.1 Fragment Growingp. 296
16.3.2 Fragment Linkingp. 297
16.3.3 Fragment Assemblyp. 299
16.4 Screening and Identification of Fragmentsp. 300
16.5 X-ray Crystallography for Fragment-based Lead Identificationp. 301
16.6 NMR Spectroscopyp. 302
16.6.1 Protein-based Methods: Structure-activity Relationship by NMRp. 302
16.6.2 Ligand-based Methodsp. 303
16.7 Mass Spectrometryp. 306
16.7.1 Covalent Mass Spectrometric Methodsp. 306
16.7.2 Non-covalent Mass Spectrometric Methodsp. 307
16.7.3 Looking at the Protein or the Ligandp. 308
16.8 Thermal Shiftp. 309
16.9 Isothermal Titration Calorimetryp. 309
16.10 Surface Plasmon Resonancep. 310
16.11 Concluding Remarksp. 311
Acknowledgementsp. 311
Referencesp. 311
Chapter 17 A Link Means a Lot: Disulfide Tethering in Structure-Based Drug DesignJeanne A. Hardy
17.1 Introduction: What is Disulfide Tethering?p. 319
17.2 Success of Native Cysteine Tetheringp. 323
17.3 Role of Structure in Engineered-cysteine Tetheringp. 325
17.4 Cooperative Tetheringp. 328
17.5 Extended Tetheringp. 330
17.6 Breakaway Tetheringp. 333
17.7 Discovery of Novel Allosteric Sites with Tetheringp. 335
17.8 Tethering as a Validation Toolp. 339
17.9 Tethering vs. Traditional Medicinal Chemistryp. 340
17.10 Tethering in Structural Determinationp. 341
17.11 The Challenge of Covalencyp. 342
17.12 Hydrophobic Bindersp. 343
17.13 Conclusions: The Future of Tetheringp. 344
Referencesp. 345
Chapter 18 The Impact of Protein Kinase Structures on Drug DiscoveryChao Zhang and Sung-Hou Kim
18.1 Introductionp. 349
18.2 The Hinge Region and the Concept of Kinase Inhibitor Scaffoldp. 351
18.3 High-throughput Crystallography for the Discovery of Novel Scaffoldsp. 353
18.3.1 High Potency-High Specificity-High Molecular (H3) Weight Screeningp. 353
18.3.2 Low Potency-Low Specificity-Low Molecular Weight (L3) Screeningp. 354
18.4 The Gatekeeper Residue and the Selectivity Pocketp. 355
18.5 The Conformational States of the DFG Motif and the Opening of the Back Pocketp. 357
18.6 Allosteric Inhibitors, Non-ATP Competitive Inhibitors, and Irreversible Inhibitorsp. 359
18.7 Discovering Kinase Inhibitors in a 500-Dimensional Spacep. 360
Acknowledgementp. 361
Referencesp. 361
Subject Indexp. 366