51
|
The influence of the 'organizational factor' on compound quality in drug discovery. Nat Rev Drug Discov 2011; 10:749-65. [DOI: 10.1038/nrd3552] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
52
|
Delvare C, Harris CS, Hennequin L, Koza P, Lambert-van der Brempt C, Pelleter J, Willerval O. Efficient three-step one-pot synthesis of a novel 2,3,5-substituted pyrazine library. ACS COMBINATORIAL SCIENCE 2011; 13:449-52. [PMID: 21648463 DOI: 10.1021/co200062n] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The partnership between rational synthesis design and mass-triggered preparative LCMS is a powerful one, capable of furnishing very large libraries in a selective manner in a very short space of time. Herein, we communicate one example of possibly a perfect marriage between the synthetic chemistry and the subsequent purification method employed, affording a ∼1000-member library supplying 50 mg on average of final compound in less than a month.
Collapse
Affiliation(s)
- Christian Delvare
- AstraZeneca, Centre de Recherches, Z.I. la Pompelle, BP1050, 51689 Reims Cedex
| | - Craig S. Harris
- AstraZeneca, Centre de Recherches, Z.I. la Pompelle, BP1050, 51689 Reims Cedex
| | - Laurent Hennequin
- AstraZeneca, Centre de Recherches, Z.I. la Pompelle, BP1050, 51689 Reims Cedex
| | - Patrice Koza
- AstraZeneca, Centre de Recherches, Z.I. la Pompelle, BP1050, 51689 Reims Cedex
| | | | - Jacques Pelleter
- AstraZeneca, Centre de Recherches, Z.I. la Pompelle, BP1050, 51689 Reims Cedex
| | - Olivier Willerval
- AstraZeneca, Centre de Recherches, Z.I. la Pompelle, BP1050, 51689 Reims Cedex
| |
Collapse
|
53
|
Cheng Y, Judd TC, Bartberger MD, Brown J, Chen K, Fremeau RT, Hickman D, Hitchcock SA, Jordan B, Li V, Lopez P, Louie SW, Luo Y, Michelsen K, Nixey T, Powers TS, Rattan C, Sickmier EA, St Jean DJ, Wahl RC, Wen PH, Wood S. From fragment screening to in vivo efficacy: optimization of a series of 2-aminoquinolines as potent inhibitors of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1). J Med Chem 2011; 54:5836-57. [PMID: 21707077 DOI: 10.1021/jm200544q] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Using fragment-based screening of a focused fragment library, 2-aminoquinoline 1 was identified as an initial hit for BACE1. Further SAR development was supported by X-ray structures of BACE1 cocrystallized with various ligands and molecular modeling studies to expedite the discovery of potent compounds. These strategies enabled us to integrate the C-3 side chain on 2-aminoquinoline 1 extending deep into the P2' binding pocket of BACE1 and enhancing the ligand's potency. We were able to improve the BACE1 potency to subnanomolar range, over 10(6)-fold more potent than the initial hit (900 μM). Further elaboration of the physical properties of the lead compounds to those more consistent with good blood-brain barrier permeability led to inhibitors with greatly improved cellular activity and permeability. Compound 59 showed an IC(50) value of 11 nM on BACE1 and cellular activity of 80 nM. This compound was advanced into rat pharmacokinetic and pharmacodynamic studies and demonstrated significant reduction of Aβ levels in cerebrospinal fluid (CSF).
Collapse
Affiliation(s)
- Yuan Cheng
- Chemistry Research and Discovery, Amgen Inc., One Amgen Center Drive, Thousand Oaks, California 91320, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Kulkarni SS, Hu X, Doi K, Wang HG, Manetsch R. Screening of protein-protein interaction modulators via sulfo-click kinetic target-guided synthesis. ACS Chem Biol 2011; 6:724-32. [PMID: 21506574 DOI: 10.1021/cb200085q] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Kinetic target-guided synthesis (TGS) and in situ click chemistry are among unconventional discovery strategies having the potential to streamline the development of protein-protein interaction modulators (PPIMs). In kinetic TGS and in situ click chemistry, the target is directly involved in the assembly of its own potent, bidentate ligand from a pool of reactive fragments. Herein, we report the use and validation of kinetic TGS based on the sulfo-click reaction between thio acids and sulfonyl azides as a screening and synthesis platform for the identification of high-quality PPIMs. Starting from a randomly designed library consisting of 9 thio acids and 9 sulfonyl azides leading to 81 potential acylsulfonamides, the target protein, Bcl-X(L), selectively assembled four PPIMs, acylsulfonamides SZ4TA2, SZ7TA2, SZ9TA1, and SZ9TA5, which have been shown to modulate Bcl-X(L)/BH3 interactions. To further investigate the Bcl-X(L) templation effect, control experiments were carried out using two mutants of Bcl-X(L). In one mutant, phenylalanine Phe131 and aspartic acid Asp133, which are critical for the BH3 domain binding, were substituted by alanines, while arginine Arg139, a residue identified to play a crucial role in the binding of ABT-737, a BH3 mimetic, was replaced by an alanine in the other mutant. Incubation of these mutants with the reactive fragments and subsequent LC/MS-SIM analysis confirmed that these building block combinations yield the corresponding acylsulfonamides at the BH3 binding site, the actual "hot spot" of Bcl-X(L). These results validate kinetic TGS using the sulfo-click reaction as a valuable tool for the straightforward identification of high-quality PPIMs.
Collapse
Affiliation(s)
- Sameer S. Kulkarni
- Department of Chemistry, University of South Florida, CHE 205, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| | - Xiangdong Hu
- Department of Chemistry, University of South Florida, CHE 205, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| | - Kenichiro Doi
- Department of Pharmacology and Penn State Hershey Cancer Institute, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
| | - Hong-Gang Wang
- Department of Pharmacology and Penn State Hershey Cancer Institute, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
| | - Roman Manetsch
- Department of Chemistry, University of South Florida, CHE 205, 4202 E. Fowler Avenue, Tampa, Florida 33620, United States
| |
Collapse
|
55
|
Congreve M, Langmead CJ, Mason JS, Marshall FH. Progress in structure based drug design for G protein-coupled receptors. J Med Chem 2011; 54:4283-311. [PMID: 21615150 PMCID: PMC3308205 DOI: 10.1021/jm200371q] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Indexed: 12/12/2022]
Affiliation(s)
- Miles Congreve
- Heptares Therapeutics Limited, BioPark, Welwyn Garden City, Hertfordshire, UK.
| | | | | | | |
Collapse
|
56
|
Sun C, Petros AM, Hajduk PJ. Fragment-based lead discovery: challenges and opportunities. J Comput Aided Mol Des 2011; 25:607-10. [DOI: 10.1007/s10822-011-9451-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 06/17/2011] [Indexed: 01/28/2023]
|
57
|
Roughley SD, Hubbard RE. How Well Can Fragments Explore Accessed Chemical Space? A Case Study from Heat Shock Protein 90. J Med Chem 2011; 54:3989-4005. [DOI: 10.1021/jm200350g] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | - Roderick E. Hubbard
- Vernalis (R&D) Ltd., Granta Park, Abington, Cambridge, CB21 6GB, U.K
- York Structural Biology Laboratory and Hull York Medical School, University of York, Heslington, York, YO10 5DD, U.K
| |
Collapse
|
58
|
Lau WF, Withka JM, Hepworth D, Magee TV, Du YJ, Bakken GA, Miller MD, Hendsch ZS, Thanabal V, Kolodziej SA, Xing L, Hu Q, Narasimhan LS, Love R, Charlton ME, Hughes S, van Hoorn WP, Mills JE. Design of a multi-purpose fragment screening library using molecular complexity and orthogonal diversity metrics. J Comput Aided Mol Des 2011; 25:621-36. [PMID: 21604056 DOI: 10.1007/s10822-011-9434-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 05/06/2011] [Indexed: 11/26/2022]
Abstract
Fragment Based Drug Discovery (FBDD) continues to advance as an efficient and alternative screening paradigm for the identification and optimization of novel chemical matter. To enable FBDD across a wide range of pharmaceutical targets, a fragment screening library is required to be chemically diverse and synthetically expandable to enable critical decision making for chemical follow-up and assessing new target druggability. In this manuscript, the Pfizer fragment library design strategy which utilized multiple and orthogonal metrics to incorporate structure, pharmacophore and pharmacological space diversity is described. Appropriate measures of molecular complexity were also employed to maximize the probability of detection of fragment hits using a variety of biophysical and biochemical screening methods. In addition, structural integrity, purity, solubility, fragment and analog availability as well as cost were important considerations in the selection process. Preliminary analysis of primary screening results for 13 targets using NMR Saturation Transfer Difference (STD) indicates the identification of uM-mM hits and the uniqueness of hits at weak binding affinities for these targets.
Collapse
Affiliation(s)
- Wan F Lau
- Pfizer Global Research and Development (PGRD), Groton, CT 06340, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Fukunishi Y. Prediction of Positions of Active Compounds Makes It Possible To Increase Activity in Fragment-Based Drug Development. Pharmaceuticals (Basel) 2011. [PMCID: PMC4055877 DOI: 10.3390/ph4050758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
We have developed a computational method that predicts the positions of active compounds, making it possible to increase activity as a fragment evolution strategy. We refer to the positions of these compounds as the active position. When an active fragment compound is found, the following lead generation process is performed, primarily to increase activity. In the current method, to predict the location of the active position, hydrogen atoms are replaced by small side chains, generating virtual compounds. These virtual compounds are docked to a target protein, and the docking scores (affinities) are examined. The hydrogen atom that gives the virtual compound with good affinity should correspond to the active position and it should be replaced to generate a lead compound. This method was found to work well, with the prediction of the active position being 2 times more efficient than random synthesis. In the current study, 15 examples of lead generation were examined. The probability of finding active positions among all hydrogen atoms was 26%, and the current method accurately predicted 60% of the active positions.
Collapse
Affiliation(s)
- Yoshifumi Fukunishi
- Biomedicinal Information Research Center (BIRC), National Institute of Advanced Industrial Science and Technology (AIST)/ 2-3-26, Aomi, Koto-ku, Tokyo 135-0064, Japan; E-Mail: ; Tel.: +81-3-3599-8290; Fax: +81-3-3599-8099
- Pharmaceutical Innovation Value Chain, BioGrid Center Kansai/ 1-4-2 Shinsenri-Higashimachi, Toyonaka, Osaka 560-0082, Japan
| |
Collapse
|
60
|
An efficient and information-rich biochemical method design for fragment library screening on ion channels. Biotechniques 2011; 49:822-9. [PMID: 21091447 DOI: 10.2144/000113538] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Drug discovery requires a simple, rapid, and cost-effective method for the early identification of novel leads and elimination of poor candidates. Here we present an experimental design that fulfils these criteria, using a ligand-gated ion channel expressed in a mammalian cell line, whose function can be probed using a voltage-sensitive dye. The experimental design is novel, as it uses the same screen to identify hit fragments and to characterize them as agonists or antagonists. The results were independently validated using radioligand binding, although the new technique has several advantages over radioligand methods. A number of novel high-affinity ligands were found. The method is broadly applicable to a wide range of receptor types including ligand-gated ion channels (LGICs), voltage-gated ion channels (VGICs), and G protein-coupled receptors (GPCRs), all of which are important drug targets.
Collapse
|
61
|
Abstract
A successful fragment-based lead discovery (FBLD) campaign largely depends on the content of the fragment collection being screened. To design a successful fragment collection, several factors must be considered, including collection size, property filters, hit follow-up considerations, and screening methods. In this chapter, we will discuss each factor and how it was applied to the design and assembly of one or more fragment collections in a major pharmaceutical company setting. We will also present examples and statistics of screening results from such collections and how subsequent collections can be improved. Lastly, we will provide a summary comparison of selected fragment collections from literature.
Collapse
|
62
|
Fragment Screening of Stabilized G-Protein-Coupled Receptors Using Biophysical Methods. Methods Enzymol 2011; 493:115-36. [DOI: 10.1016/b978-0-12-381274-2.00005-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
63
|
Congreve M, Langmead C, Marshall FH. The use of GPCR structures in drug design. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2011; 62:1-36. [PMID: 21907905 DOI: 10.1016/b978-0-12-385952-5.00011-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Structure-based drug discovery is routinely applied to soluble targets such as proteases and kinases. It is only recently that multiple high-resolution X-ray structures of G protein-coupled receptors (GPCRs) have become available. Here we review the technology developments that have led to the recent plethora of GPCR structures. These include developments in protein expression and purification as well as techniques to stabilize receptors and crystallize them. We discuss the findings derived from the new structures with regard to understanding GPCR function and pharmacology. Finally, we examine the utility of structure-based drug discovery approaches including homology modeling, virtual screening, and fragment screening for GPCRs in the context of what has been learnt from other target classes.
Collapse
Affiliation(s)
- Miles Congreve
- Heptares Therapeutics, Biopark, Welwyn Garden City, Hertfordshire, United Kingdom
| | | | | |
Collapse
|
64
|
Computational medicinal chemistry in fragment-based drug discovery: what, how and when. Future Med Chem 2011; 3:95-134. [DOI: 10.4155/fmc.10.277] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The use of fragment-based drug discovery (FBDD) has increased in the last decade due to the encouraging results obtained to date. In this scenario, computational approaches, together with experimental information, play an important role to guide and speed up the process. By default, FBDD is generally considered as a constructive approach. However, such additive behavior is not always present, therefore, simple fragment maturation will not always deliver the expected results. In this review, computational approaches utilized in FBDD are reported together with real case studies, where applicability domains are exemplified, in order to analyze them, and then, maximize their performance and reliability. Thus, a proper use of these computational tools can minimize misleading conclusions, keeping the credit on FBDD strategy, as well as achieve higher impact in the drug-discovery process. FBDD goes one step beyond a simple constructive approach. A broad set of computational tools: docking, R group quantitative structure–activity relationship, fragmentation tools, fragments management tools, patents analysis and fragment-hopping, for example, can be utilized in FBDD, providing a clear positive impact if they are utilized in the proper scenario – what, how and when. An initial assessment of additive/non-additive behavior is a critical point to define the most convenient approach for fragments elaboration.
Collapse
|
65
|
Davies DR, Begley DW, Hartley RC, Staker BL, Stewart LJ. Predicting the Success of Fragment Screening by X-Ray Crystallography. Methods Enzymol 2011; 493:91-114. [DOI: 10.1016/b978-0-12-381274-2.00004-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
66
|
Abstract
Fragment-based drug discovery (FBDD) has emerged in the past decade as a powerful tool for discovering drug leads. The approach first identifies starting points: very small molecules (fragments) that are about half the size of typical drugs. These fragments are then expanded or linked together to generate drug leads. Although the origins of the technique date back some 30 years, it was only in the mid-1990s that experimental techniques became sufficiently sensitive and rapid for the concept to be become practical. Since that time, the field has exploded: FBDD has played a role in discovery of at least 18 drugs that have entered the clinic, and practitioners of FBDD can be found throughout the world in both academia and industry. Literally dozens of reviews have been published on various aspects of FBDD or on the field as a whole, as have three books (Jahnke and Erlanson, Fragment-based approaches in drug discovery, 2006; Zartler and Shapiro, Fragment-based drug discovery: a practical approach, 2008; Kuo, Fragment based drug design: tools, practical approaches, and examples, 2011). However, this chapter will assume that the reader is approaching the field with little prior knowledge. It will introduce some of the key concepts, set the stage for the chapters to follow, and demonstrate how X-ray crystallography plays a central role in fragment identification and advancement.
Collapse
|
67
|
|
68
|
Brötz-Oesterhelt H, Sass P. Postgenomic strategies in antibacterial drug discovery. Future Microbiol 2010; 5:1553-79. [DOI: 10.2217/fmb.10.119] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
During the last decade the field of antibacterial drug discovery has changed in many aspects including bacterial organisms of primary interest, discovery strategies applied and pharmaceutical companies involved. Target-based high-throughput screening had been disappointingly unsuccessful for antibiotic research. Understanding of this lack of success has increased substantially and the lessons learned refer to characteristics of targets, screening libraries and screening strategies. The ‘genomics’ approach was replaced by a diverse array of discovery strategies, for example, searching for new natural product leads among previously abandoned compounds or new microbial sources, screening for synthetic inhibitors by targeted approaches including structure-based design and analyses of focused libraries and designing resistance-breaking properties into antibiotics of established classes. Furthermore, alternative treatment options are being pursued including anti-virulence strategies and immunotherapeutic approaches. This article summarizes the lessons learned from the genomics era and describes discovery strategies resulting from that knowledge.
Collapse
Affiliation(s)
- Heike Brötz-Oesterhelt
- AiCuris, Wuppertal, Germany, Institute for Pharmaceutical Biology, University of Duesseldorf, Universitätsstrasse 1, Building 26.23.U1, Germany
| | - Peter Sass
- Institute of Medical Microbiology, Immunology & Parasitology, Pharmaceutical Microbiology Section, University of Bonn, Germany
| |
Collapse
|
69
|
Boettcher A, Ruedisser S, Erbel P, Vinzenz D, Schiering N, Hassiepen U, Rigollier P, Mayr LM, Woelcke J. Fragment-Based Screening by Biochemical Assays. ACTA ACUST UNITED AC 2010; 15:1029-41. [DOI: 10.1177/1087057110380455] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fragment-based screening (FBS) has gained acceptance in the pharmaceutical industry as an attractive approach for the identification of new chemical starting points for drug discovery programs in addition to classical strategies such as high-throughput screening. There is the concern that screening of fragments at high µM concentrations in biochemical assays results in increased false-positive and false-negative rates. Here the authors systematically compare the data quality of FBS obtained by enzyme activity-based fluorescence intensity, fluorescence lifetime, and mobility shift assays with the data quality from surface plasmon resonance (SPR) and nuclear magnetic resonance (NMR) methods. The serine protease trypsin and the matrix metalloprotease MMP12 were selected as model systems. For both studies, 352 fragments were selected each. From the data generated, all 3 biochemical protease assay methods can be used for screening of fragments with low false-negative and low false-positive rates, comparable to those achieved with the SPR-based assays. It can also be concluded that only fragments with a solubility higher than the screening concentration determined by means of NMR should be used for FBS purposes. Extrapolated to 10,000 fragments, the biochemical assays speed up the primary FBS process by approximately a factor of 10 and reduce the protease consumption by approximately 10,000-fold compared to NMR protein observation experiments.
Collapse
Affiliation(s)
- Andreas Boettcher
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Simon Ruedisser
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Paulus Erbel
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Daniela Vinzenz
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Nikolaus Schiering
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Ulrich Hassiepen
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Pascal Rigollier
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Lorenz M. Mayr
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| | - Julian Woelcke
- Novartis Institutes for BioMedical Research (NIBR), Expertise Platform Proteases (EPP), Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
70
|
Oyarzabal J, Zarich N, Albarran MI, Palacios I, Urbano-Cuadrado M, Mateos G, Reymundo I, Rabal O, Salgado A, Corrionero A, Fominaya J, Pastor J, Bischoff JR. Discovery of Mitogen-Activated Protein Kinase-Interacting Kinase 1 Inhibitors by a Comprehensive Fragment-Oriented Virtual Screening Approach. J Med Chem 2010; 53:6618-28. [DOI: 10.1021/jm1005513] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Julen Oyarzabal
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Natasha Zarich
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - María Isabel Albarran
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Irene Palacios
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Manuel Urbano-Cuadrado
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Genoveva Mateos
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Isabel Reymundo
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Obdulia Rabal
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Antonio Salgado
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Ana Corrionero
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Jesús Fominaya
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Joaquin Pastor
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - James R. Bischoff
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
71
|
Hubbard RE. Structure-based drug discovery and protein targets in the CNS. Neuropharmacology 2010; 60:7-23. [PMID: 20673774 DOI: 10.1016/j.neuropharm.2010.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Accepted: 07/21/2010] [Indexed: 12/18/2022]
Abstract
Structure-based methods are having an increasing role and impact in drug discovery. The crystal structures of an increasing number of therapeutic targets are becoming available. These structures can transform our understanding of how these proteins perform their biological function and often provide insights into the molecular basis of disease. In addition, the structures can help the discovery process. Methods such as virtual screening and experimental fragment screening can provide starting hit compounds for a discovery project. Crystal structures of compounds bound to the protein can direct or guide the medicinal chemistry optimisation to improve drug-like properties - not only providing ideas on how to improve binding affinity or selectivity, but also showing where the compound can be modified in attempting to modulate physico-chemical properties and biological efficacy. The majority of drug discovery projects against globular protein targets now use these methods at some stage. This review provides a summary of the range of structure-based drug discovery methods that are in use and surveys the suitability of the methods for targets currently identified for CNS drugs. Until recently, structure-based discovery was difficult or unknown for these targets. The recent determination of the structures of a number of GPCR proteins, together with the steady increase in structures for other membrane proteins, is opening up the possibility for these structure-based methods to find increased use in drug discovery for CNS diseases and conditions.
Collapse
|
72
|
Holdgate GA, Anderson M, Edfeldt F, Geschwindner S. Affinity-based, biophysical methods to detect and analyze ligand binding to recombinant proteins: matching high information content with high throughput. J Struct Biol 2010; 172:142-57. [PMID: 20609391 DOI: 10.1016/j.jsb.2010.06.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 06/28/2010] [Accepted: 06/29/2010] [Indexed: 01/21/2023]
Abstract
Affinity-based technologies have become impactful tools to detect, monitor and characterize molecular interactions using recombinant target proteins. This can aid the understanding of biological function by revealing mechanistic details, and even more importantly, enables the identification of new improved ligands that can modulate the biological activity of those targets in a desired fashion. The selection of the appropriate technology is a key step in that process, as each one of the currently available technologies offers a characteristic type of biophysical information about the ligand-binding event. Alongside the indisputable advantages of each of those technologies they naturally display diverse restrictions that are quite frequently related to the target system to be studied but also to the affinity, solubility and molecular size of the ligands. This paper discusses some of the theoretical and experimental aspects of the most common affinity-based methods, what type of information can be gained from each one of those approaches, and what requirements as well as limitations are expected from working with recombinant proteins on those platforms and how those can be optimally addressed.
Collapse
Affiliation(s)
- Geoff A Holdgate
- Lead Generation Sciences, AstraZeneca R&D Alderley Park, Mereside, Alderley Park, United Kingdom
| | | | | | | |
Collapse
|
73
|
Abstract
Nuclear magnetic resonance (NMR) is a versatile technique for the pharmaceutical industry. From organic chemistry to MRI, there are a number of applications of NMR. Among them, biomolecular NMR has been used for structure determination of biomolecules and analyzing the interaction between a target protein and its inhibitors. In the context of fragment-based drug discovery (FBDD), NMR has been known as a fragment screening technique, because NMR is good at detecting a weak binding compound in an accurate manner. Generally, the NMR technique for fragment screening is classified into two families: the ligand-based technique and the protein-based technique. The latter technique requires stable isotope labeled protein and also can be applied to a relatively small MW protein target. In the ligand-based technique such as saturation transfer difference (STD) and WaterLOGSY, only the NMR signals of the ligands are observed. The disadvantage of STD and WaterLOGSY is that the non-specific binding is also observed and a competition experiment is required in order to select the specific binding compound. Due to the difference in the consumption of the protein sample, the ligand-based technique has generally been used recently as a primary screening.
Collapse
Affiliation(s)
- Hiroyuki Hanzawa
- Exploratory Research Laboratories I, Daiichi Sankyo Co., Ltd, Japan.
| | | |
Collapse
|
74
|
Drewry DH, Macarron R. Enhancements of screening collections to address areas of unmet medical need: an industry perspective. Curr Opin Chem Biol 2010; 14:289-98. [PMID: 20413343 DOI: 10.1016/j.cbpa.2010.03.024] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 02/19/2010] [Accepted: 03/24/2010] [Indexed: 01/31/2023]
Abstract
The past 20 years have witnessed an impressive expansion of the 'drug space'; defined as the intersection of the Medicinal Chemistry space and the Biologically Active space relevant in the quest for new treatments for disease. Despite the success of known lead discovery tactics, areas of unmet medical need are often linked to challenging or novel targets and are poorly served by current screening collections. A successful strategy to fill the gaps is to diversify the approaches taken in the enhancement of screening collections. Possible strategies include investments through proven methods, exploring areas of chemical space previously neglected (e.g. hydrophilic compounds, natural product mimics), and applying tactics to the lead discovery process that are complementary to HTS (e.g. fragment based screening or multidisciplinary team efforts to tackle new target classes).
Collapse
Affiliation(s)
- David H Drewry
- Molecular Discovery Research, GlaxoSmithKline, 5 Moore Drive, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
75
|
Ward RA. Using protein-ligand docking to assess the chemical tractability of inhibiting a protein target. J Mol Model 2010; 16:1833-43. [DOI: 10.1007/s00894-010-0683-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 01/31/2010] [Indexed: 11/24/2022]
|
76
|
Boyd SM, de Kloe GE. Fragment library design: efficiently hunting drugs in chemical space. DRUG DISCOVERY TODAY. TECHNOLOGIES 2010; 7:e147-e202. [PMID: 24103769 DOI: 10.1016/j.ddtec.2010.11.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
|
77
|
Whittaker M, Law RJ, Ichihara O, Hesterkamp T, Hallett D. Fragments: past, present and future. DRUG DISCOVERY TODAY. TECHNOLOGIES 2010; 7:e147-e202. [PMID: 24103768 DOI: 10.1016/j.ddtec.2010.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
|
78
|
Congreve M, Marshall F. The impact of GPCR structures on pharmacology and structure-based drug design. Br J Pharmacol 2009; 159:986-96. [PMID: 19912230 DOI: 10.1111/j.1476-5381.2009.00476.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
After many years of effort, recent technical breakthroughs have enabled the X-ray crystal structures of three G-protein-coupled receptors (GPCRs) (beta1 and beta2 adrenergic and adenosine A(2a)) to be solved in addition to rhodopsin. GPCRs, like other membrane proteins, have lagged behind soluble drug targets such as kinases and proteases in the number of structures available and the level of understanding of these targets and their interaction with drugs. The availability of increasing numbers of structures of GPCRs is set to greatly increase our understanding of some of the key issues in GPCR biology. In particular, what constitutes the different receptor conformations that are involved in signalling and the molecular changes which occur upon receptor activation. How future GPCR structures might alter our views on areas such as agonist-directed signalling and allosteric regulation as well as dimerization is discussed. Knowledge of crystal structures in complex with small molecules will enable techniques in drug discovery and design, which have previously only been applied to soluble targets, to now be used for GPCR targets. These methods include structure-based drug design, virtual screening and fragment screening. This review considers how these methods have been used to address problems in drug discovery for kinase and protease targets and therefore how such methods are likely to impact GPCR drug discovery in the future.
Collapse
Affiliation(s)
- Miles Congreve
- Heptares Therapeutics Ltd, Welwyn Garden City, Hertfordshire, UK
| | | |
Collapse
|
79
|
Orita M, Warizaya M, Amano Y, Ohno K, Niimi T. Advances in fragment-based drug discovery platforms. Expert Opin Drug Discov 2009; 4:1125-44. [DOI: 10.1517/17460440903317580] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
80
|
Vulpetti A, Hommel U, Landrum G, Lewis R, Dalvit C. Design and NMR-Based Screening of LEF, a Library of Chemical Fragments with Different Local Environment of Fluorine. J Am Chem Soc 2009; 131:12949-59. [DOI: 10.1021/ja905207t] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Anna Vulpetti
- Novartis Institute for Biomedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| | - Ulrich Hommel
- Novartis Institute for Biomedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| | - Gregory Landrum
- Novartis Institute for Biomedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| | - Richard Lewis
- Novartis Institute for Biomedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| | - Claudio Dalvit
- Novartis Institute for Biomedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
81
|
Shapiro AB, Walkup GK, Keating TA. Correction for Interference by Test Samples in High-Throughput Assays. ACTA ACUST UNITED AC 2009; 14:1008-16. [DOI: 10.1177/1087057109341768] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In high-throughput biochemical assays performed in multiwell plates, the effect of test samples on the activity of the biochemical system is usually measured by optical means such as absorbance, fluorescence, luminescence, or scintillation counting. The test sample often causes detection interference when it remains in the well during the measurement. Interference may be due to light absorption, fluorescence quenching, sample fluorescence, chemical interaction of the sample with a detection reagent, or depression of the meniscus. A simple method is described that corrects for such interference well by well. The interference is measured in a separate artifact assay plate. An appropriate arithmetic correction is then applied to the measurement in the corresponding well of the activity assay plate. The correction procedure can be used for single-point screening or potency measurements on serial dilutions of test samples.
Collapse
|
82
|
|
83
|
Challenges of fragment screening. J Comput Aided Mol Des 2009; 23:449-51. [DOI: 10.1007/s10822-009-9293-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 06/10/2009] [Indexed: 10/20/2022]
|
84
|
The multiple roles of computational chemistry in fragment-based drug design. J Comput Aided Mol Des 2009; 23:459-73. [PMID: 19533374 DOI: 10.1007/s10822-009-9284-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 05/18/2009] [Indexed: 10/20/2022]
Abstract
Fragment-based drug discovery (FBDD) represents a change in strategy from the screening of molecules with higher molecular weights and physical properties more akin to fully drug-like compounds, to the screening of smaller, less complex molecules. This is because it has been recognised that fragment hit molecules can be efficiently grown and optimised into leads, particularly after the binding mode to the target protein has been first determined by 3D structural elucidation, e.g. by NMR or X-ray crystallography. Several studies have shown that medicinal chemistry optimisation of an already drug-like hit or lead compound can result in a final compound with too high molecular weight and lipophilicity. The evolution of a lower molecular weight fragment hit therefore represents an attractive alternative approach to optimisation as it allows better control of compound properties. Computational chemistry can play an important role both prior to a fragment screen, in producing a target focussed fragment library, and post-screening in the evolution of a drug-like molecule from a fragment hit, both with and without the available fragment-target co-complex structure. We will review many of the current developments in the area and illustrate with some recent examples from successful FBDD discovery projects that we have conducted.
Collapse
|
85
|
Chen IJ, Hubbard RE. Lessons for fragment library design: analysis of output from multiple screening campaigns. J Comput Aided Mol Des 2009; 23:603-20. [PMID: 19495994 DOI: 10.1007/s10822-009-9280-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Accepted: 05/07/2009] [Indexed: 11/26/2022]
Abstract
Over the past 8 years, we have developed, refined and applied a fragment based discovery approach to a range of protein targets. Here we report computational analyses of various aspects of our fragment library and the results obtained for fragment screening. We reinforce the finding of others that the experimentally observed hit rate for screening fragments can be related to a computationally defined druggability index for the target. In general, the physicochemical properties of the fragment hits display the same profile as the library, as is expected for a truly diverse library which probes the relevant chemical space. An analysis of the fragment hits against various protein classes has shown that the physicochemical properties of the fragments are complementary to the properties of the target binding site. The effectiveness of some fragments appears to be achieved by an appropriate mix of pharmacophore features and enhanced aromaticity, with hydrophobic interactions playing an important role. The analysis emphasizes that it is possible to identify small fragments that are specific for different binding sites. To conclude, we discuss how the results could inform further development and improvement of our fragment library.
Collapse
Affiliation(s)
- I-Jen Chen
- Vernalis (R&D) Ltd, Granta Park, Cambridge, CB21 6GB, UK
| | | |
Collapse
|
86
|
Virtual fragment screening: an exploration of various docking and scoring protocols for fragments using Glide. J Comput Aided Mol Des 2009; 23:527-39. [DOI: 10.1007/s10822-009-9281-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 05/07/2009] [Indexed: 10/20/2022]
|
87
|
Perspicace S, Banner D, Benz J, Müller F, Schlatter D, Huber W. Fragment-Based Screening Using Surface Plasmon Resonance Technology. ACTA ACUST UNITED AC 2009; 14:337-49. [DOI: 10.1177/1087057109332595] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Surface plasmon resonance (SPR) technology has emerged as a new and powerful technique to investigate the interaction between low-molecular-weight molecules and target proteins. In the present work, the authors assemble from a large compound collection a library of 2226 molecules (fragments having low molecular weights between 100 and 300 Da) to screen them for binding to chymase, a serine protease. Both the active chymase and a zymogen-like form of the protein were used in parallel to distinguish between specific and unspecific binding. The relative ligand-binding activity of the immobilized protein was periodically measured with a reference compound. The screening experiments were performed at 25 °C at a fragment concentration of 200 µM in the presence of 2% DMSO. Applying the filter cascade, affinity—selectivity—competition (competition with reference compounds and cross-competition with fragments), 80 compounds show up as positive screening hits. Competition experiments between fragments show that they bind to different parts of the active site. Of 36 fragments co-crystallized for X-ray studies, 12 could be located in the active site of the protein. These results validate the authors' library and demonstrate that the application of SPR technology as a filter in fragment screening can be achieved successfully. ( Journal of Biomolecular Screening. 2009:337-349)
Collapse
Affiliation(s)
- Samantha Perspicace
- F. Hoffmann-La Roche Ltd, Pharma Research, Discovery Technologies, Basel, Switzerland,
| | - David Banner
- F. Hoffmann-La Roche Ltd, Pharma Research, Discovery Technologies, Basel, Switzerland
| | - Jörg Benz
- F. Hoffmann-La Roche Ltd, Pharma Research, Discovery Technologies, Basel, Switzerland
| | - Francis Müller
- F. Hoffmann-La Roche Ltd, Pharma Research, Discovery Technologies, Basel, Switzerland
| | - Daniel Schlatter
- F. Hoffmann-La Roche Ltd, Pharma Research, Discovery Technologies, Basel, Switzerland
| | - Walter Huber
- F. Hoffmann-La Roche Ltd, Pharma Research, Discovery Technologies, Basel, Switzerland
| |
Collapse
|
88
|
Fukunishi Y, Mashimo T, Orita M, Ohno K, Nakamura H. In Silico Fragment Screening by Replica Generation (FSRG) Method for Fragment-Based Drug Design. J Chem Inf Model 2009; 49:925-33. [DOI: 10.1021/ci800435x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yoshifumi Fukunishi
- Biomedicinal Information Research Center (BIRC), National Institute of Advanced Industrial Science and Technology (AIST), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan, Pharmaceutical Innovation Value Chain, BioGrid Center Kansai, 1-4-2 Shinsenri-Higashimachi, Toyonaka, Osaka 560-0082, Japan, Information and Mathematical Science Laboratory Inc., Meikei Building, 1-5-21, Ohtsuka, Bunkyo-ku, Tokyo, 112-0012, Japan, Japan Biological Informatics Consortium (JBIC), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan,
| | - Tadaaki Mashimo
- Biomedicinal Information Research Center (BIRC), National Institute of Advanced Industrial Science and Technology (AIST), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan, Pharmaceutical Innovation Value Chain, BioGrid Center Kansai, 1-4-2 Shinsenri-Higashimachi, Toyonaka, Osaka 560-0082, Japan, Information and Mathematical Science Laboratory Inc., Meikei Building, 1-5-21, Ohtsuka, Bunkyo-ku, Tokyo, 112-0012, Japan, Japan Biological Informatics Consortium (JBIC), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan,
| | - Masaya Orita
- Biomedicinal Information Research Center (BIRC), National Institute of Advanced Industrial Science and Technology (AIST), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan, Pharmaceutical Innovation Value Chain, BioGrid Center Kansai, 1-4-2 Shinsenri-Higashimachi, Toyonaka, Osaka 560-0082, Japan, Information and Mathematical Science Laboratory Inc., Meikei Building, 1-5-21, Ohtsuka, Bunkyo-ku, Tokyo, 112-0012, Japan, Japan Biological Informatics Consortium (JBIC), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan,
| | - Kazuki Ohno
- Biomedicinal Information Research Center (BIRC), National Institute of Advanced Industrial Science and Technology (AIST), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan, Pharmaceutical Innovation Value Chain, BioGrid Center Kansai, 1-4-2 Shinsenri-Higashimachi, Toyonaka, Osaka 560-0082, Japan, Information and Mathematical Science Laboratory Inc., Meikei Building, 1-5-21, Ohtsuka, Bunkyo-ku, Tokyo, 112-0012, Japan, Japan Biological Informatics Consortium (JBIC), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan,
| | - Haruki Nakamura
- Biomedicinal Information Research Center (BIRC), National Institute of Advanced Industrial Science and Technology (AIST), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan, Pharmaceutical Innovation Value Chain, BioGrid Center Kansai, 1-4-2 Shinsenri-Higashimachi, Toyonaka, Osaka 560-0082, Japan, Information and Mathematical Science Laboratory Inc., Meikei Building, 1-5-21, Ohtsuka, Bunkyo-ku, Tokyo, 112-0012, Japan, Japan Biological Informatics Consortium (JBIC), 2-41-6, Aomi, Koto-ku, Tokyo 135-0064, Japan,
| |
Collapse
|
89
|
de Kloe GE, Bailey D, Leurs R, de Esch IJP. Transforming fragments into candidates: small becomes big in medicinal chemistry. Drug Discov Today 2009; 14:630-46. [PMID: 19443265 DOI: 10.1016/j.drudis.2009.03.009] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 03/12/2009] [Accepted: 03/16/2009] [Indexed: 11/15/2022]
Abstract
Fragment-based drug discovery (FBDD) represents a logical and efficient approach to lead discovery and optimisation. It can draw on structural, biophysical and biochemical data, incorporating a wide range of inputs, from precise mode-of-binding information on specific fragments to wider ranging pharmacophoric screening surveys using traditional HTS approaches. It is truly an enabling technology for the imaginative medicinal chemist. In this review, we analyse a representative set of 23 published FBDD studies that describe how low molecular weight fragments are being identified and efficiently transformed into higher molecular weight drug candidates. FBDD is now becoming warmly endorsed by industry as well as academia and the focus on small interacting molecules is making a big scientific impact.
Collapse
Affiliation(s)
- Gerdien E de Kloe
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
90
|
Blomberg N, Cosgrove DA, Kenny PW, Kolmodin K. Design of compound libraries for fragment screening. J Comput Aided Mol Des 2009; 23:513-25. [DOI: 10.1007/s10822-009-9264-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2008] [Accepted: 02/16/2009] [Indexed: 01/15/2023]
|
91
|
Holloway MK, Hunt P, McGaughey GB. Structure and modeling in the design of β- and γ-secretase inhibitors. Drug Dev Res 2009. [DOI: 10.1002/ddr.20291] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
92
|
Orita M, Ohno K, Niimi T. Two ‘Golden Ratio’ indices in fragment-based drug discovery. Drug Discov Today 2009; 14:321-8. [DOI: 10.1016/j.drudis.2008.10.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 10/07/2008] [Accepted: 10/22/2008] [Indexed: 10/21/2022]
|
93
|
Tanrikulu Y, Schneider G. Pseudoreceptor models in drug design: bridging ligand- and receptor-based virtual screening. Nat Rev Drug Discov 2008; 7:667-77. [DOI: 10.1038/nrd2615] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
94
|
Hesterkamp T, Whittaker M. Fragment-based activity space: smaller is better. Curr Opin Chem Biol 2008; 12:260-8. [PMID: 18316043 DOI: 10.1016/j.cbpa.2008.02.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 01/29/2008] [Accepted: 02/02/2008] [Indexed: 12/18/2022]
Abstract
Fragment-based drug discovery has the potential to supersede traditional high throughput screening based drug discovery for molecular targets amenable to structure determination. This is because the chemical diversity coverage is better accomplished by a fragment collection of reasonable size than by larger HTS collections. Furthermore, fragments have the potential to be efficient target binders with higher probability than more elaborated drug-like compounds. The selection of the fragment screening technique is driven by sensitivity and throughput considerations, and we advocate in the present article the use of high concentration bioassays in conjunction with NMR-based hit confirmation. Subsequent ligand X-ray structure determination of the fragment ligand in complex with the target protein by co-crystallisation or crystal soaking can focus on confirmed binders.
Collapse
|
95
|
Perrin D, Frémaux C, Scheer A. Assay Development and Screening of a Serine/Threonine Kinase in an On-Chip Mode Using Caliper Nanofluidics Technology. ACTA ACUST UNITED AC 2006; 11:359-68. [PMID: 16751332 DOI: 10.1177/1087057106286653] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Kinases are key targets for drug discovery. In the field of screening in general and especially in the kinase area, because of considerations of efficiency and cost, radioactivity-based assays tend to be replaced by alternative, mostly fluorescence-based, assays. Today, the limiting factor is rarely the number of data points that can be obtained but rather the quality of the data, enzyme availability, and cost. In this article, the authors describe the development of an assay for a kinase screen based on the electrophoretic separation of fluorescent product and substrate using a Caliper-based nanofluidics environment in on-chip incubation mode. The authors present the results of screening a focused set of 32,000 compounds together with confirmation data obtained in a filtration assay. In addition, they have made a small-scale comparison between the on-chip and off-chip nanofluidics screening modes. In their hands, the screen in on-chip mode is characterized by high precision most likely due to the absence of liquid pipetting; an excellent confirmation rate (62%) in an independent assay format, namely, filtration; and good sensitivity. This study led to the identification of 4 novel chemical series of inhibitors.
Collapse
Affiliation(s)
- Dominique Perrin
- Molecular Screening and Cellular Pharmacology Department, Serono Pharmaceutical Research Institute, Geneva, Switzerland.
| | | | | |
Collapse
|