Details

Small Molecule Medicinal Chemistry


Small Molecule Medicinal Chemistry

Strategies and Technologies
1. Aufl.

von: Werngard Czechtizky, Peter Hamley

132,99 €

Verlag: Wiley
Format: EPUB
Veröffentl.: 25.09.2015
ISBN/EAN: 9781118771693
Sprache: englisch
Anzahl Seiten: 528

DRM-geschütztes eBook, Sie benötigen z.B. Adobe Digital Editions und eine Adobe ID zum Lesen.

Beschreibungen

Stressing strategic and technological solutions to medicinal chemistry challenges, this book presents methods and practices for optimizing the chemical aspects of drug discovery. Chapters discuss benefits, challenges, case studies, and industry perspectives for improving drug discovery programs with respect to quality and costs.<br /><br />•    Focuses on small molecules and their critical role in medicinal chemistry, reviewing chemical and economic advantages, challenges, and trends in the field from industry perspectives<br />•    Discusses novel approaches and key topics, like screening collection enhancement, risk sharing, HTS triage, new lead finding approaches, diversity-oriented synthesis, peptidomimetics, natural products, and high throughput medicinal chemistry approaches<br />•    Explains how to reduce design-make-test cycle times by integrating medicinal chemistry, physical chemistry, and ADME profiling techniques<br />•    Includes descriptive case studies, examples, and applications to illustrate new technologies and provide step-by-step explanations to enable them in a laboratory setting
<p>List of Contributors xiii</p> <p>Introduction 1<br /><i>Werngard Czechtizky and Peter Hamley</i></p> <p><b>Part I Exploring Biological Space: Access to New Collections 11</b></p> <p><b>1 Elements for the Development of Strategies for Compound Library Enhancement 13<br /></b><i>Edgar Jacoby</i></p> <p>1.1 Introduction 13</p> <p>1.2 Chemical Space for Drug Discovery 14</p> <p>1.3 Molecular Properties for Drug Discovery 17</p> <p>1.4 Major Compound Classes 21</p> <p>1.5 Chemical Design Approaches to Expand Bioactive Chemical Space 25</p> <p>1.6 Conclusion 28</p> <p>Acknowledgments 29</p> <p>References 29</p> <p><b>2 The European Lead Factory 37<br /></b><i>Christopher Kallus, Jörg Hüser, Philip S. Jones, and Adam Nelson</i></p> <p>2.1 Introduction 37</p> <p>2.1.1 Background 37</p> <p>2.1.2 The European Lead Factory 38</p> <p>2.2 Building the Joint European Compound Library 43</p> <p>2.2.1 Definition of Criteria and an Approach for the Review and Selection of Library Proposals 46</p> <p>2.2.2 Collation, Review, and Selection of an Initial Wave of Library Proposals 47</p> <p>2.2.3 A Web?]Based Tool to Support the Collation, Review, and Selection of Proposals 49</p> <p>2.2.4 Synthetic Validation of Library Proposals and Library Production 49</p> <p>2.3 Qualified Hit Generation 54</p> <p>2.3.1 Capabilities of the ESC 54</p> <p>2.3.2 Target Selection and Generation of Qualified Hits 56</p> <p>2.3.3 Exploitation of Qualified Hit List 58</p> <p>2.4 Future Perspectives 58</p> <p>Acknowledgments 59</p> <p>References 59</p> <p><b>3 Access to Compound Collections: New Business Models for Compound Acquisition and Sharing 61<br /></b><i>Peter ten Holte</i></p> <p>3.1 Introduction 61</p> <p>3.1.1 Vertical Disintegration and the Quest for Innovation 61</p> <p>3.1.2 Innovative Chemistry 63</p> <p>3.1.3 Access to Supplementary Compound Collections 63</p> <p>3.2 Risk?]Sharing Approaches 64</p> <p>3.2.1 Overview 64</p> <p>3.2.2 Blinded Screening 65</p> <p>3.2.3 Follow?]Up of Blinded Screening: Various Models 65</p> <p>3.3 Library Exchange 69</p> <p>3.3.1 Partners with Different Scientific Interests 70</p> <p>3.3.2 Partners with Similar Scientific Interests 70</p> <p>3.3.3 Compound Selection: Use and Potential Risks 71</p> <p>3.4 Sharing Collections for External Screening 72</p> <p>3.4.1 Rationale 72</p> <p>3.4.2 Academic Drug Discovery Consortium (Addc) 72</p> <p>3.4.3 Eu?]Openscreen 73</p> <p>3.4.4 N IH Roadmap 73</p> <p>3.5 Conclusion 74</p> <p>Acknowledgments 74</p> <p>References 75</p> <p><b>Part II Exploring Biological Space: Access to New Chemistries 77</b></p> <p><b>4 New Advances in Diversity?]Oriented Synthesis 79<br /></b><i>Warren R. J. D. Galloway, Jamie E. Stokes, and David R. Spring</i></p> <p>4.1 Introduction: Small Molecules and Biology 79</p> <p>4.2 The Need for Structural Diversity in Synthetic Small Molecule Screening Collections 80</p> <p>4.3 Diversity?]Oriented Synthesis of New Structurally Diverse Compound Collections 82</p> <p>4.3.1 General Principles of Diversity?]Oriented Synthesis 82</p> <p>4.3.2 Achieving Structural Diversity: The Importance of Scaffold Diversity 83</p> <p>4.3.3 Synthetic Principles in DOS 83</p> <p>4.3.4 Scaffold Diversity and Molecular Type 86</p> <p>4.3.5 Examples of DOS Campaigns 86</p> <p>4.4 Concluding Remarks 97</p> <p>References 98</p> <p><b>5 Solid?]Phase Combinatorial Chemistry 103<br /></b><i>Marcel Patek, Martin Smrcina, Eric Wegrzyniak, Victor Nikolaev, and Andres Mariscal</i></p> <p>5.1 Introduction 103</p> <p>5.2 Chapter Outline 104</p> <p>5.3 Combinatorial Chemistry in Retrospect 104</p> <p>5.4 Foundations of Solid?]Phase Synthesis of Combinatorial Chemistry 107</p> <p>5.4.1 Ingredients of Solid?]Phase Chemistry 109</p> <p>5.4.2 Library Development and Production 117</p> <p>5.4.3 Analytical Chemistry and Solid?]Phase Synthesis of Libraries 129</p> <p>5.5 The Outcome of Tucson Combinatorial Chemistry at Sanofi 132</p> <p>5.5.1 Overall Strategy 132</p> <p>5.5.2 Drug Discovery Outcomes 134</p> <p>5.5.3 Key Parameters of Combichem Productivity 134</p> <p>5.6 Conclusions and Outlook 135</p> <p>References 136</p> <p><b>6 Recent Advances in Multicomponent Reaction Chemistry: Applications in Small Molecule Drug Discovery 145</b><br /><i>Christopher Hulme, Muhammad Ayaz, Guillermo Martinez?]Ariza, Federico Medda, and Arthur Shaw</i></p> <p>6.1 Introduction 145</p> <p>6.2 Classical Multi-Component Reactions (MCRs) 147</p> <p>6.3 The Passerini Reaction (Mario Passerini 1921) 147</p> <p>6.4 Ugi Reaction 147</p> <p>6.4.1 The Ugi-deprotect-cyclize (UDC) strategy 152</p> <p>6.4.2 Bi-functional approach (BIFA) 153</p> <p>6.4.3 Miscellaneous Post?]Ugi Condensations 154</p> <p>6.5 Van Leusen Reaction 154</p> <p>6.6 Petasis Reaction 155</p> <p>6.7 Groebke–Blackburn–Bienaymé (GBB) Reaction 155</p> <p>6.8 Recently Discovered Novel MCRs 155</p> <p>6.8.1 Cyclic Anhydride?]Based MCRs 155</p> <p>6.8.2 1?]Azadiene?]Based MCRs 156</p> <p>6.8.3 Recent IMCRs and Secondary Reactions 157</p> <p>6.8.4 Miscellaneous MCRs 159</p> <p>6.9 Asymmetric MCRs 159</p> <p>6.10 Applications of MCRs in Medicinal Chemistry 160</p> <p>6.10.1 Kinase Inhibitors 161</p> <p>6.10.2 Protease Inhibitors 163</p> <p>6.10.3 Ion Channel Inhibitors 165</p> <p>6.10.4 Protein–Protein Interaction Inhibitors 165</p> <p>6.10.5 Tubulin Polymerization Inhibitors 166</p> <p>6.10.6 G?]Protein?]Coupled Receptors 168</p> <p>6.11 Summary 171</p> <p>References 171</p> <p>Part III Screening Strategies 189</p> <p><b>7 Computational Techniques to Support Hit Triage 191<br /></b><i>Douglas B. Kitchen and Hélène Y. Decornez</i></p> <p>7.1 Lead Finding Process: Overview and Challenges 191</p> <p>7.1.1 The Need for Triage 191</p> <p>7.1.2 The Lead Generation Process 191</p> <p>7.1.3 Hit Triage: From Actives to Hits to Hit Series 193</p> <p>7.1.4 Challenges to Successful Lead Finding 194</p> <p>7.1.5 Frequent Hitters 195</p> <p>7.1.6 Implications of Human Decision?]Making 195</p> <p>7.2 Chemical Structure Analysis of Hit Lists 196</p> <p>7.2.1 Similarity?]Based Clustering 197</p> <p>7.2.2 Scaffold?]Based Clustering 198</p> <p>7.2.3 Application of Clustering Classification Methods 201</p> <p>7.3 Rules and Filters 201</p> <p>7.3.1 Computational Descriptors for Property Assessment 202</p> <p>7.3.2 Lipophilicity and Other Physicochemical Descriptors 205</p> <p>7.3.3 Structural and Shape Descriptors 205</p> <p>7.3.4 Multiparameter Calculations: MPO and QED 206</p> <p>7.3.5 Frequent?]Hitter Analysis 207</p> <p>7.3.6 Reactive Group Analysis 209</p> <p>7.4 Triage Systems 210</p> <p>7.5 Ligand Efficiency Indices 210</p> <p>7.6 Hit Series Analysis 211</p> <p>7.6.1 Latent Hit Series and Singletons 211</p> <p>7.6.2 Rapid Hit Exploration and Compound Set Enrichment 211</p> <p>7.6.3 SAR Analysis 212</p> <p>7.6.4 Data Volume, Integration, Retrieval, and Visualization 213</p> <p>7.7 Summary 214</p> <p>References 214</p> <p><b>8 Fragment?]Based Drug Discovery 221<br /></b><i>Jean?]Paul Renaud, Thomas Neumann, and Luc Van Hijfte</i></p> <p>8.1 Introduction 221</p> <p>8.2 Fragment Libraries 223</p> <p>8.3 Biophysical Screening Technologies 223</p> <p>8.3.1 Surface Plasmon Resonance (SPR) 224</p> <p>8.3.2 Nuclear Magnetic Resonance (NMR) 231</p> <p>8.3.3 X?]Ray Crystallography 234</p> <p>8.3.4 Noncovalent Mass Spectrometry 235</p> <p>8.3.5 Differential Scanning Fluorimetry (DSF) 237</p> <p>8.3.6 Biophysical Techniques for Fragment Screening against Membrane Proteins 238</p> <p>8.3.7 Biophysical Techniques for Fragment Screening against PPIs 238</p> <p>8.4 Fragment Evolution Strategies 239</p> <p>8.5 Fbdd Case Studies 240</p> <p>8.5.1 Aurora Kinase Inhibitors 240</p> <p>8.5.2 Tackling PPIs: Fragment?]Based Discovery of Bromodomain Inhibitor Leads 241</p> <p>8.6 The Future 243</p> <p>References 244</p> <p><b>9 Virtual Screening 251<br /></b><i>Karl?]Heinz Baringhaus and Gerhard Hessler</i></p> <p>9.1 Introduction 251</p> <p>9.1.1 Goals of Virtual Screening 252</p> <p>9.2 Databases and Database Preparation 254</p> <p>9.3 Validation of the Virtual Screening Strategy 256</p> <p>9.4 Ligand?]Based Virtual Screening 258</p> <p>9.4.1 2D Approaches 259</p> <p>9.4.2 3D Ligand?]Based Approaches 261</p> <p>9.5 Structure?]Based Virtual Screening 263</p> <p>9.6 Other Virtual Screening Applications 266</p> <p>9.7 Conclusion 268</p> <p>References 269</p> <p><b>10 Phenotypic Screening 281<br /></b><i>Michelle Palmer</i></p> <p>10.1 Introduction 281</p> <p>10.2 History and Past Successes 282</p> <p>10.3 Impact of Phenotypic Screening 282</p> <p>10.4 Model Systems for Phenotypic Assays 285</p> <p>10.4.1 Cell Lines 285</p> <p>10.4.2 Primary and Stem Cells 285</p> <p>10.4.3 Cocultures 286</p> <p>10.4.4 3D Cell Models 287</p> <p>10.5 Assays 287</p> <p>10.5.1 Assay Technologies 287</p> <p>10.5.2 Assay Development Considerations 290</p> <p>10.5.3 Example 1: Selective Killing of Breast Cancer Stem Cells 291</p> <p>10.5.4 Example 2: CFTR Potentiator Drug 291</p> <p>10.6 Deorphaning 292</p> <p>10.6.1 Affinity?]Based Proteomics 292</p> <p>10.6.2 Genetic Profiling 295</p> <p>10.6.3 Target Profiling 296</p> <p>10.6.4 Comodifier Profiling 296</p> <p>10.6.5 Target Engagement 297</p> <p>10.6.6 Example 3: Elucidating MOA for a Regulator of Polyploidization 297</p> <p>10.7 Summary 298</p> <p>References 299</p> <p><b>Part IV Technologies for Medicinal Chemistry Optimization 305</b></p> <p><b>11 Advances in the Understanding of Drug Properties in Medicinal Chemistry 307<br /></b><i>Peter Hamley and Patrick Jimonet</i></p> <p>11.1 Introduction 307</p> <p>11.2 Properties and Origins of Marketed Drugs 308</p> <p>11.2.1 The Consistent Properties of Oral Drugs 308</p> <p>11.2.2 The Changing Origins of Oral Drugs 308</p> <p>11.3 Drug Properties and Attrition in Clinical Development 310</p> <p>11.4 The Rule of Five 312</p> <p>11.4.1 The Concept 312</p> <p>11.4.2 Druggability 313</p> <p>11.5 The Concept of Lead?]Likeness 313</p> <p>11.5.1 The Consequences on Screening and Collections 314</p> <p>11.6 Influence of Drug Properties on Absorption, Distribution, Metabolism, Excretion, and Toxicity 314</p> <p>11.7 Building on the Ro5: New Guidelines for Compound Design 316</p> <p>11.7.1 Ligand Efficiency 316</p> <p>11.7.2 Ligand Lipophilicity Efficiency and Other Indices 317</p> <p>11.7.3 Chemical Beauty 318</p> <p>11.8 Alternatives, Criticisms, and Exceptions 318</p> <p>11.9 Conclusions 320</p> <p>References 320</p> <p><b>12 Recent Developments in Automated Solution Phase Library Production 323<br /></b><i>Thomas C. Maier and Werngard Czechtizky</i></p> <p>12.1 Introduction 323</p> <p>12.1.1 Introduction and Definitions 323</p> <p>12.1.2 Library Types 324</p> <p>12.1.3 Chemotypes 326</p> <p>12.2 Library Production 327</p> <p>12.2.1 The Library Production Process 327</p> <p>12.2.2 Process Optimization 330</p> <p>12.3 New Technologies in Automated Liquid?]Phase Library Synthesis 334</p> <p>12.3.1 Provision of Starting Materials: Automated Reagent Dispensaries 334</p> <p>12.3.2 Microwave 335</p> <p>12.3.3 Library Purification: Automated RP?]HPLC and SFC as Orthogonal Methods 336</p> <p>12.4 Flow Chemistry and Gas?]Phase Reactions 342</p> <p>12.4.1 Reactive Gases in Flow 344</p> <p>12.5 Conclusion 345</p> <p>References 345</p> <p><b>13 A DME Profiling: An Introduction for the Medicinal Chemist 353<br /></b><i>Katharina Mertsch, Martin Will, Werngard Czechtizky, Niels Griesang, Alexander Marker, and Jacob Olsen</i></p> <p>13.1 Introduction 353</p> <p>13.2 Compound Profiling in H2L Optimization 354</p> <p>13.2.1 Intestinal Absorption 354</p> <p>13.2.2 Drug Metabolism and Inhibition of CYP450 Enzymes 355</p> <p>13.2.3 Protein Binding 356</p> <p>13.2.4 En Route to a Lead Series: In Vivo PK Studies 358</p> <p>13.3 Compound Profiling in Lead Optimization 359</p> <p>13.3.1 Extended CYP Inhibition Studies 359</p> <p>13.3.2 Mechanism?]Based CYP Inhibition 359</p> <p>13.3.3 Inhibition of Transport Proteins 360</p> <p>13.3.4 Biopharmaceutical Classification of a Clinical Candidate (Classification of Potential Drugs into Biopharmaceutical Classification System or Biopharmaceutical Drug Disposition and Classification System) 360</p> <p>13.4 Integration of Medicinal Chemistry, Biology, Physicochemical, and ADME Profiling: Strategies Toward Cycle Time Reductions 362</p> <p>13.4.1 Planning Phase 363</p> <p>13.4.2 Sample Preparation and Distribution 364</p> <p>13.4.3 Compound QC 365</p> <p>13.4.4 Determination of Physicochemical Properties 367</p> <p>13.4.5 ADME Profiling: General Remarks 369</p> <p>13.4.6 Metabolic Lability Profiling 369</p> <p>13.4.7 Permeability Testing 370</p> <p>13.4.8 CYP Inhibition Profiling 372</p> <p>13.5 Summary 372</p> <p>References 373</p> <p><b>Part V Medicinal Chemistry beyond Small Molecules 379</b></p> <p><b>14 The Role of Natural Products in Drug Discovery: Examples of Marketed Drugs 381<br /></b><i>Lars Ole Haustedt and Karsten Siems</i></p> <p>14.1 Natural Products and Natural Product Derivatives in Commercial Drugs 381</p> <p>14.2 Hit to Lead Optimization of Natural Product Hits 397</p> <p>14.3 Case Study 1: Taxol 397</p> <p>14.4 Case Study 2: Epothilone 406</p> <p>14.5 Case Study 3: Eribulin 407</p> <p>14.6 Case Study 4: Geldanamycin 413</p> <p>14.7 Case Study 5: Ingenol Mebutate (Picato) 417</p> <p>14.8 Summary 422</p> <p>References 423</p> <p><b>15 Peptidomimetics of α?]Helical and β?]Strand Protein Binding Epitopes 431<br /></b><i>Nina Bionda and Rudi Fasan</i></p> <p>15.1 Protein–Protein Interactions as Therapeutic Targets 431</p> <p>15.2 Peptidomimetics of α?]Helical Protein Binding Epitopes 433</p> <p>15.2.1 α?]Helix?]Mediated PPIs 433</p> <p>15.2.2 Side?]Chain Cross?]Linked α?]Helices 435</p> <p>15.2.3 Hydrogen?]Bond Surrogate?]Stabilized α?]Helices 442</p> <p>15.2.4 Other Type I α?]Helix Peptidomimetics 443</p> <p>15.2.5 Type III α?]Helix Peptidomimetics 445</p> <p>15.3 Peptidomimetics of β?]Strand Protein Binding Epitopes 446</p> <p>15.3.1 β?]Strand?]Mediated PPIs 446</p> <p>15.3.2 Type I β?]Strand Peptidomimetics 447</p> <p>15.3.3 Type III β?]Strand Peptidomimetics 449</p> <p>15.4 Conclusion 452</p> <p>References 453</p> <p><b>16 In Vivo Imaging of Drug Action 465<br /></b><i>Oliver Plettenburg and Matthias Löhn</i></p> <p>16.1 Introduction 465</p> <p>16.2 Overview of Imaging Methods 466</p> <p>16.2.1 Fluorescence?]Based Methods 466</p> <p>16.2.2 MRI 470</p> <p>16.2.3 CT 470</p> <p>16.2.4 PET/SPECT 471</p> <p>16.3 Imaging of Therapeutic Effects 476</p> <p>16.3.1 Cancer 476</p> <p>16.3.2 Diabetes 483</p> <p>16.3.3 CNS Disorders 486</p> <p>16.4 Conclusion and Outlook 490</p> <p>References 491</p> <p>Index 503</p> <p> </p>
<p>"This book describes a large variety of methodologies that are employed in drug discovery and early development. The subjects covered range from the design and administration of chemical libraries, access to new chemical scaffolds and chemical diversity, to characterization of target binding and ADME profiling methods. While the book focuses on traditional small-molecule medicinal chemistry, an outlook to non-traditional targets such as protein–protein interactions and imaging applications is also provided. Most of the authors have a background in the pharmaceutical industry. The book therefore allows an insight into laboratories that are less prone to publish their experiences than their academic counterparts, and the trends and observations which accumulate over the years. In addition, particularly the academic reader is presented with sometimes refreshingly independent views on certain popular topics in medicinal chemistry and chemical biology that have reached “nuisance” status in recent years. For example, the discussion on “hit triage” contains a wide range of literature reference points beyond the currently over-debated assay-interference compounds that will allow the interested reader to obtain a well-balanced view. For an academic who is heavily involved in teaching duties, the book offers numerous highly useful overviews, such as the development chart of combinatorial chemistry methods. Although the heydays of combi-chem have long passed, the concept has remained an important addition to the toolbox of medicinal</p> <p>chemistry, also in academic laboratories. Fittingly, one of the following chapters provides a review of multicomponent</p> <p>reactions. This chapter is filled with numerous highly appreciated figures that collect and demonstrate the broad chemical diversity which is accessible via traditional and recently developed multicomponent reactions, a real treasure chest that will certainly be useful in teaching and research. There are some weaker parts in the book. This is, for example, the natural</p> <p>products chapter, which starts with a reference to a “famous”, highly biased publication that “proves” the importance of</p> <p>natural products by classifying, for example, purely synthetic ATP-competitive kinase inhibitors as somehow natural-product- derived. This is followed by a long table that is mostly, and not surprisingly, composed of antibiotics and steroids of semisynthetic origin. Next, however, appear some interesting examples of such semi-synthetic strategies, which will be highly instructive for students and practitioners of medicinal chemistry. And these latter examples clearly demonstrate that it is actually not necessary to make natural products appear more important by creating just the right definition for terms such as “natural-product-derived”. Somewhat annoying is the use of an insert for color figures, requiring frequent “lookups”, particularly since the captions are not provided along with the color figures. However, this technical aspect is really minor, and may have been necessary to keep the price of the book at a reasonable level—about £130, which could also be in the range of interested students. For the reviewer, selected fragments of the book will certainly be integrated into the teaching curriculum, and the book will be recommended for students at all levels. "<br /><b>(Prof. Christian Klein Heidelberg University - ChemMedChem, July 2017)</b></p>
<p><b>Werngard Czechtizky </b>is the Head of Medicinal Chemistry of the German Hub of Sanofi, based in Frankfurt, Germany.  She has wide experience in lead generation and lead optimization for central nervous system, cardiovascular and diabetes targets, and her teams have been responsible for a number of leads and clinical candidates in these areas over the last years. She was educated at ETH Zürich and Harvard University, USA.<br /><br /><b>Peter Hamley </b>is the global head of External Innovation and Sourcing for chemistry, computational chemistry, and screening technologies at Sanofi, based in Frankfurt, Germany.  He spent ten years at AstraZeneca in the United Kingdom, and then moved to Sanofi as a medicinal chemistry leader, building their automated chemistry capabilities and natural product technology. He was educated at Imperial College, London, the University of Cambridge and the University of Pennsylvania.</p>
Small molecules are the major source for marketed therapeutic drugs and valuable tools for studying biological pathways. To identify new small molecule therapeutics in an increasingly competitive environment, researchers need to keep up-to-date with the rapidly changing world of drug discovery.<br /><br />This is where <i>Small Molecule Medicinal Chemistry: Strategies and Technologies</i> comes in, helping medicinal chemists and colleagues in related disciplines to identify and progress new innovative small molecules as efficiently as possible. <br /><br />This book provides an overview of the field, with a focus on recent trends and up-to-date technologies for the identification and development of small molecule drugs. The all-inclusive set of successful techniques and methods is divided into 5 parts: access to new compound collections, new chemistries, lead identification strategies, technologies for medicinal chemistry optimization, and an overview of the chemical space beyond small molecules.<br /><br />Stressing strategic and technological solutions to medicinal chemistry challenges, this book presents methods and practices for optimizing the chemical aspects of drug discovery. Among the key features, <i>Small Molecule Medicinal Chemistry</i>:<br /><br />•    Discusses benefits, challenges, case studies, and industry perspectives for improving drug discovery programs in terms of quality and costs<br />•    Focuses on small molecules and their critical role in medicinal chemistry, reviewing chemical and economic advantages and challenges, as well as trends in the field from an industry perspective<br />•    Discusses novel approaches and key topics, like screening collection enhancement, risk sharing, HTS triage, new lead finding approaches, diversity-oriented synthesis, peptidomimetics, natural products, and high throughput medicinal chemistry approaches

Diese Produkte könnten Sie auch interessieren:

Hot-Melt Extrusion
Hot-Melt Extrusion
von: Dennis Douroumis
PDF ebook
136,99 €
Hot-Melt Extrusion
Hot-Melt Extrusion
von: Dennis Douroumis
EPUB ebook
136,99 €
Kunststoffe
Kunststoffe
von: Wilhelm Keim
PDF ebook
99,99 €