1
|
Zheng G, Wu D, Wei X, Xu D, Mao T, Yan D, Han W, Shang X, Chen Z, Qiu J, Tang K, Cao Z, Qiu T. PbsNRs: predict the potential binders and scaffolds for nuclear receptors. Brief Bioinform 2024; 26:bbae710. [PMID: 39798999 PMCID: PMC11724720 DOI: 10.1093/bib/bbae710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025] Open
Abstract
Nuclear receptors (NRs) are a class of essential proteins that regulate the expression of specific genes and are associated with multiple diseases. In silico methods for prescreening potential NR binders with predictive binding ability are highly desired for NR-related drug development but are rarely reported. Here, we present the PbsNRs (Predicting binders and scaffolds for Nuclear Receptors), a user-friendly web server designed to predict the potential NR binders and scaffolds through proteochemometric modeling. The utility of PbsNRs was systemically evaluated using both chemical compounds and natural products. Results indicated that PbsNRs achieved a good prediction performance for chemical compounds on internal (ROC-AUC = 0.906, where ROC is Receiver-Operating Characteristic curve and AUC is the Area Under the Curve) and external (ROC-AUC = 0.783) datasets, outperforming both compound-ligand interaction tools and NR-specific predictors. PbsNRs also successfully identified bioactive chemical scaffolds for NRs by screening massive natural products. Moreover, the predicted bioactive and inactive natural products for NR2B1 were experimentally validated using biosensors. PbsNRs not only aids in screening potential therapeutic NR binders but also helps discover the essential molecular scaffold and guide the drug discovery for multiple NR-related diseases. The PbsNRs web server is available at http://pbsnrs.badd-cao.net.
Collapse
Affiliation(s)
- Genhui Zheng
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
- Oden Institute for Computational Engineering and Sciences (ICES), University of Texas at Austin, No. 201 East 24th Street, Austin 78712, TX, United States
| | - Dingfeng Wu
- National Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou 310052, China
| | - Xiuxia Wei
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Dongpo Xu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Tiantian Mao
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Deyu Yan
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Wenhao Han
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Xiaoxiao Shang
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
- Department of Mathematics and Statistics, McGill University, 805 Sherbrooke Street West, Montreal H3A 0B9, Quebec, Canada
| | - Zikun Chen
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Jingxuan Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Road, Yangpu District, Shanghai 200093, China
| | - Kailin Tang
- School of Life Sciences and Technology, Tongji University, No. 1239 Siping Road, Shanghai 200092, China
| | - Zhiwei Cao
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
| | - Tianyi Qiu
- Institute of Clinical Science, Zhongshan Hospital, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Intelligent Medicine Institute, School of Life Sciences, Fudan University, No. 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
2
|
Cichońska A, Ravikumar B, Rahman R. AI for targeted polypharmacology: The next frontier in drug discovery. Curr Opin Struct Biol 2024; 84:102771. [PMID: 38215530 DOI: 10.1016/j.sbi.2023.102771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/30/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024]
Abstract
In drug discovery, targeted polypharmacology, i.e., targeting multiple molecular targets with a single drug, is redefining therapeutic design to address complex diseases. Pre-selected pharmacological profiles, as exemplified in kinase drugs, promise enhanced efficacy and reduced toxicity. Historically, many of such drugs were discovered serendipitously, limiting predictability and efficacy, but currently artificial intelligence (AI) offers a transformative solution. Machine learning and deep learning techniques enable modeling protein structures, generating novel compounds, and decoding their polypharmacological effects, opening an avenue for more systematic and predictive multi-target drug design. This review explores the use of AI in identifying synergistic co-targets and delineating them from anti-targets that lead to adverse effects, and then discusses advances in AI-enabled docking, generative chemistry, and proteochemometric modeling of proteome-wide compound interactions, in the context of polypharmacology. We also provide insights into challenges ahead.
Collapse
|
3
|
Gorostiola González M, van den Broek RL, Braun TGM, Chatzopoulou M, Jespers W, IJzerman AP, Heitman LH, van Westen GJP. 3DDPDs: describing protein dynamics for proteochemometric bioactivity prediction. A case for (mutant) G protein-coupled receptors. J Cheminform 2023; 15:74. [PMID: 37641107 PMCID: PMC10463931 DOI: 10.1186/s13321-023-00745-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/10/2023] [Indexed: 08/31/2023] Open
Abstract
Proteochemometric (PCM) modelling is a powerful computational drug discovery tool used in bioactivity prediction of potential drug candidates relying on both chemical and protein information. In PCM features are computed to describe small molecules and proteins, which directly impact the quality of the predictive models. State-of-the-art protein descriptors, however, are calculated from the protein sequence and neglect the dynamic nature of proteins. This dynamic nature can be computationally simulated with molecular dynamics (MD). Here, novel 3D dynamic protein descriptors (3DDPDs) were designed to be applied in bioactivity prediction tasks with PCM models. As a test case, publicly available G protein-coupled receptor (GPCR) MD data from GPCRmd was used. GPCRs are membrane-bound proteins, which are activated by hormones and neurotransmitters, and constitute an important target family for drug discovery. GPCRs exist in different conformational states that allow the transmission of diverse signals and that can be modified by ligand interactions, among other factors. To translate the MD-encoded protein dynamics two types of 3DDPDs were considered: one-hot encoded residue-specific (rs) and embedding-like protein-specific (ps) 3DDPDs. The descriptors were developed by calculating distributions of trajectory coordinates and partial charges, applying dimensionality reduction, and subsequently condensing them into vectors per residue or protein, respectively. 3DDPDs were benchmarked on several PCM tasks against state-of-the-art non-dynamic protein descriptors. Our rs- and ps3DDPDs outperformed non-dynamic descriptors in regression tasks using a temporal split and showed comparable performance with a random split and in all classification tasks. Combinations of non-dynamic descriptors with 3DDPDs did not result in increased performance. Finally, the power of 3DDPDs to capture dynamic fluctuations in mutant GPCRs was explored. The results presented here show the potential of including protein dynamic information on machine learning tasks, specifically bioactivity prediction, and open opportunities for applications in drug discovery, including oncology.
Collapse
Affiliation(s)
- Marina Gorostiola González
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- ONCODE Institute, Leiden, The Netherlands
| | - Remco L van den Broek
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Thomas G M Braun
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Magdalini Chatzopoulou
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Willem Jespers
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- ONCODE Institute, Leiden, The Netherlands
| | - Gerard J P van Westen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
4
|
Smilova MD, Curran PR, Radoux CJ, von Delft F, Cole JC, Bradley AR, Marsden BD. Fragment Hotspot Mapping to Identify Selectivity-Determining Regions between Related Proteins. J Chem Inf Model 2022; 62:284-294. [PMID: 35020376 PMCID: PMC8790751 DOI: 10.1021/acs.jcim.1c00823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
Selectivity is a
crucial property in small molecule development.
Binding site comparisons within a protein family are a key piece of
information when aiming to modulate the selectivity profile of a compound.
Binding site differences can be exploited to confer selectivity for
a specific target, while shared areas can provide insights into polypharmacology.
As the quantity of structural data grows, automated methods are needed
to process, summarize, and present these data to users. We present
a computational method that provides quantitative and data-driven
summaries of the available binding site information from an ensemble
of structures of the same protein. The resulting ensemble maps identify
the key interactions important for ligand binding in the ensemble.
The comparison of ensemble maps of related proteins enables the identification
of selectivity-determining regions within a protein family. We applied
the method to three examples from the well-researched human bromodomain
and kinase families, demonstrating that the method is able to identify
selectivity-determining regions that have been used to introduce selectivity
in past drug discovery campaigns. We then illustrate how the resulting
maps can be used to automate comparisons across a target protein family.
Collapse
Affiliation(s)
- Mihaela D Smilova
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K
| | - Peter R Curran
- The Cambridge Crystallographic Data Centre (CCDC), Cambridge CB2 1EZ, U.K.,Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Chris J Radoux
- Exscientia Ltd., The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, U.K
| | - Frank von Delft
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K.,Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot OX11 0DE, U.K.,Research Complex at Harwell. Harwell Science and Innovation Campus, Didcot OX11 0FA, U.K.,Department of Biochemistry, University of Johannesburg, Auckland Park 2006, South Africa
| | - Jason C Cole
- The Cambridge Crystallographic Data Centre (CCDC), Cambridge CB2 1EZ, U.K
| | - Anthony R Bradley
- Exscientia Ltd., The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, U.K
| | - Brian D Marsden
- Centre for Medicines Discovery, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, U.K.,Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7DQ, U.K
| |
Collapse
|
5
|
Rasti B. Quantitative Characterization of the Chemical Space Governed by Human Carbonic Anhydrases and selenium-containing derivatives of solfonamides. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
6
|
Parks C, Gaieb Z, Amaro RE. An Analysis of Proteochemometric and Conformal Prediction Machine Learning Protein-Ligand Binding Affinity Models. Front Mol Biosci 2020; 7:93. [PMID: 32671093 PMCID: PMC7328444 DOI: 10.3389/fmolb.2020.00093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/22/2020] [Indexed: 11/13/2022] Open
Abstract
Protein-ligand binding affinity is a key pharmacodynamic endpoint in drug discovery. Sole reliance on experimental design, make, and test cycles is costly and time consuming, providing an opportunity for computational methods to assist. Herein, we present results comparing random forest and feed-forward neural network proteochemometric models for their ability to predict pIC50 measurements for held out generic Bemis-Murcko scaffolds. In addition, we assess the ability of conformal prediction to provide calibrated prediction intervals in both a retrospective and semi-prospective test using the recently released Grand Challenge 4 data set as an external test set. In total, random forest and deep neural network proteochemometric models show quality retrospective performance but suffer in the semi-prospective setting. However, the conformal predictor prediction intervals prove to be well-calibrated both retrospectively and semi-prospectively showing that they can be used to guide hit discovery and lead optimization campaigns.
Collapse
Affiliation(s)
| | | | - Rommie E. Amaro
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
7
|
|
8
|
Gagic Z, Ruzic D, Djokovic N, Djikic T, Nikolic K. In silico Methods for Design of Kinase Inhibitors as Anticancer Drugs. Front Chem 2020; 7:873. [PMID: 31970149 PMCID: PMC6960140 DOI: 10.3389/fchem.2019.00873] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Rational drug design implies usage of molecular modeling techniques such as pharmacophore modeling, molecular dynamics, virtual screening, and molecular docking to explain the activity of biomolecules, define molecular determinants for interaction with the drug target, and design more efficient drug candidates. Kinases play an essential role in cell function and therefore are extensively studied targets in drug design and discovery. Kinase inhibitors are clinically very important and widely used antineoplastic drugs. In this review, computational methods used in rational drug design of kinase inhibitors are discussed and compared, considering some representative case studies.
Collapse
Affiliation(s)
- Zarko Gagic
- Department of Pharmaceutical Chemistry, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Nemanja Djokovic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Teodora Djikic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
9
|
Giblin KA, Hughes SJ, Boyd H, Hansson P, Bender A. Prospectively Validated Proteochemometric Models for the Prediction of Small-Molecule Binding to Bromodomain Proteins. J Chem Inf Model 2018; 58:1870-1888. [DOI: 10.1021/acs.jcim.8b00400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kathryn A. Giblin
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Samantha J. Hughes
- Computational Chemistry, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge CB10 1XL, U.K
| | - Helen Boyd
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg 431 50 SE, Sweden
| | - Pia Hansson
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg 431 50 SE, Sweden
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| |
Collapse
|
10
|
Rasti B, Mazraedoost S, Panahi H, Falahati M, Attar F. New insights into the selective inhibition of the β-carbonic anhydrases of pathogenic bacteria Burkholderia pseudomallei and Francisella tularensis: a proteochemometrics study. Mol Divers 2018; 23:263-273. [PMID: 30120657 DOI: 10.1007/s11030-018-9869-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/09/2018] [Indexed: 10/28/2022]
Abstract
Nowadays, antibiotic resistance has turned into one of the most important worldwide health problems. Biological end point of critical enzymes induced by potent inhibitors is recently being considered as a highly effective and popular strategy to defeat antibiotic-resistant pathogens. For instance, the simple but critical β-carbonic anhydrase has recently been in the center of attention for anti-pathogen drug discoveries. However, no β-carbonic anhydrase selective inhibitor has yet been developed. Available β-carbonic anhydrase inhibitors are also highly potent with regard to human carbonic anhydrases, leading to severe inevitable side effects in case of usage. Therefore, developing novel inhibitors with high selectivity against pathogenic β-carbonic anhydrases is of great essence. Herein, for the first time, we have conducted a proteochemometric study to explore the structural and the chemical aspects of the interactions governed by bacterial β-carbonic anhydrases and their inhibitors. We have found valuable information which can lead to designing novel inhibitors with better selectivity for bacterial β-carbonic anhydrases.
Collapse
Affiliation(s)
- Behnam Rasti
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran.
| | - Sargol Mazraedoost
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Hanieh Panahi
- Department of Mathematics and Statistics, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advance Science and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Farnoosh Attar
- Department of Biology, Faculty of Food Industry and Agriculture, Standard Research Institute (SRI), Karaj, Iran
| |
Collapse
|
11
|
Qiu T, Wu D, Qiu J, Cao Z. Finding the molecular scaffold of nuclear receptor inhibitors through high-throughput screening based on proteochemometric modelling. J Cheminform 2018; 10:21. [PMID: 29651663 PMCID: PMC5897275 DOI: 10.1186/s13321-018-0275-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 04/02/2018] [Indexed: 02/10/2023] Open
Abstract
Nuclear receptors (NR) are a class of proteins that are responsible for sensing steroid and thyroid hormones and certain other molecules. In that case, NR have the ability to regulate the expression of specific genes and associated with various diseases, which make it essential drug targets. Approaches which can predict the inhibition ability of compounds for different NR target should be particularly helpful for drug development. In this study, proteochemometric modelling was introduced to analysis the bioactivity between chemical compounds and NR targets. Results illustrated the ability of our PCM model for high-throughput NR-inhibitor screening after evaluated on both internal (AUC > 0.870) and external (AUC > 0.746) validation set. Moreover, in-silico predicted bioactive compounds were clustered according to structure similarity and a series of representative molecular scaffolds can be derived for five major NR targets. Through scaffolds analysis, those essential bioactive scaffolds of different NR target can be detected and compared. Generally, the methods and molecular scaffolds proposed in this article can not only help the screening of potential therapeutic NR-inhibitors but also able to guide the future NR-related drug discovery.
Collapse
Affiliation(s)
- Tianyi Qiu
- School of Life Sciences and Technology, Shanghai 10th People's Hospital, Tongji University, No. 1239 SiPing Road, Shanghai, China.,The Institute of Biomedical Sciences, Fudan University, No. 138 Medical College Road, Shanghai, China
| | - Dingfeng Wu
- School of Life Sciences and Technology, Shanghai 10th People's Hospital, Tongji University, No. 1239 SiPing Road, Shanghai, China
| | - Jingxuan Qiu
- School of Life Sciences and Technology, Shanghai 10th People's Hospital, Tongji University, No. 1239 SiPing Road, Shanghai, China.,School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, No. 516 JunGong Road, Shanghai, China
| | - Zhiwei Cao
- School of Life Sciences and Technology, Shanghai 10th People's Hospital, Tongji University, No. 1239 SiPing Road, Shanghai, China.
| |
Collapse
|
12
|
Rasti B, Shahangian SS. Proteochemometric modeling of the origin of thymidylate synthase inhibition. Chem Biol Drug Des 2018; 91:1007-1016. [PMID: 29251822 DOI: 10.1111/cbdd.13163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/09/2017] [Accepted: 12/01/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Behnam Rasti
- Department of Microbiology; Faculty of Basic Sciences; Lahijan Branch; Islamic Azad University (IAU); Lahijan Guilan Iran
| | | |
Collapse
|
13
|
Sorgenfrei FA, Fulle S, Merget B. Kinome-Wide Profiling Prediction of Small Molecules. ChemMedChem 2017; 13:495-499. [PMID: 28544552 DOI: 10.1002/cmdc.201700180] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/20/2017] [Indexed: 12/21/2022]
Abstract
Extensive kinase profiling data, covering more than half of the human kinome, are available nowadays and allow the construction of activity prediction models of high practical utility. Proteochemometric (PCM) approaches use compound and protein descriptors, which enables the extrapolation of bioactivity values to thus far unexplored kinases. In this study, the potential of PCM to make large-scale predictions on the entire kinome is explored, considering the applicability on novel compounds and kinases, including clinically relevant mutants. A rigorous validation indicates high predictive power on left-out kinases and superiority over individual kinase QSAR models for new compounds. Furthermore, external validation on clinically relevant mutant kinases reveals an excellent predictive power for mutations spread across the ATP binding site.
Collapse
Affiliation(s)
- Frieda A Sorgenfrei
- BioMed X Innovation Center, Im Neuenheimer Feld 515, 69120, Heidelberg, Germany
| | - Simone Fulle
- BioMed X Innovation Center, Im Neuenheimer Feld 515, 69120, Heidelberg, Germany
| | - Benjamin Merget
- BioMed X Innovation Center, Im Neuenheimer Feld 515, 69120, Heidelberg, Germany
| |
Collapse
|
14
|
Subramanian V, Ain QU, Henno H, Pietilä LO, Fuchs JE, Prusis P, Bender A, Wohlfahrt G. 3D proteochemometrics: using three-dimensional information of proteins and ligands to address aspects of the selectivity of serine proteases. MEDCHEMCOMM 2017; 8:1037-1045. [PMID: 30108817 PMCID: PMC6072133 DOI: 10.1039/c6md00701e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/14/2017] [Indexed: 11/21/2022]
Abstract
The high similarity between certain sub-pockets of serine proteases may lead to low selectivity of protease inhibitors. Therefore the application of proteochemometrics (PCM), which quantifies the relationship between protein/ligand descriptors and affinity for multiple ligands and targets simultaneously, is useful to understand and improve the selectivity profiles of potential inhibitors. In this study, protein field-based PCM that uses knowledge-based and WaterMap derived fields to describe proteins in combination with 2D (RDKit and MOE fingerprints) and 3D (4 point pharmacophoric fingerprints and GRIND) ligand descriptors was used to model the bioactivities of 24 homologous serine proteases and 5863 inhibitors in an integrated fashion. Of the multiple field-based PCM models generated based on different ligand descriptors, RDKit fingerprints showed the best performance in terms of external prediction with Rtest2 of 0.72 and RMSEP of 0.81. Further, visual interpretation of the models highlights sub-pocket specific regions that influence affinity and selectivity of serine protease inhibitors.
Collapse
Affiliation(s)
- Vigneshwari Subramanian
- Division of Pharmaceutical Chemistry and Technology , Faculty of Pharmacy , University of Helsinki , 00014 Helsinki , Finland
- Computer-Aided Drug Design , Orion Pharma , Orionintie 1 , 02101 Espoo , Finland .
| | - Qurrat Ul Ain
- Centre for Molecular Informatics , Department of Chemistry , Lensfield Road , CB2 1EW Cambridge , UK
| | - Helena Henno
- Computer-Aided Drug Design , Orion Pharma , Orionintie 1 , 02101 Espoo , Finland .
| | - Lars-Olof Pietilä
- Computer-Aided Drug Design , Orion Pharma , Orionintie 1 , 02101 Espoo , Finland .
| | - Julian E Fuchs
- Centre for Molecular Informatics , Department of Chemistry , Lensfield Road , CB2 1EW Cambridge , UK
- Institute of General , Inorganic and Theoretical Chemistry , University of Innsbruck , Innrain 82 , 6020 Innsbruck , Austria
| | - Peteris Prusis
- Computer-Aided Drug Design , Orion Pharma , Orionintie 1 , 02101 Espoo , Finland .
| | - Andreas Bender
- Centre for Molecular Informatics , Department of Chemistry , Lensfield Road , CB2 1EW Cambridge , UK
| | - Gerd Wohlfahrt
- Computer-Aided Drug Design , Orion Pharma , Orionintie 1 , 02101 Espoo , Finland .
| |
Collapse
|
15
|
Shaikh N, Sharma M, Garg P. Selective Fusion of Heterogeneous Classifiers for Predicting Substrates of Membrane Transporters. J Chem Inf Model 2017; 57:594-607. [PMID: 28228010 DOI: 10.1021/acs.jcim.6b00508] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Membrane transporters play a crucial role in determining fate of administered drugs in a biological system. Early identification of plausible transporters for a drug molecule can provide insights into its therapeutic, pharmacokinetic, and toxicological profiles. In the present study, predictive models for classifying small molecules into substrates and nonsubstrates of various pharmaceutically important membrane transporters were developed using quantitative structure-activity relationship (QSAR) and proteochemometric (PCM) approaches. For this purpose, 4575 substrate interactions for these transporters were collected from the Metabolism and Transport Database (Metrabase) and the literature. The transporters selected for this study include (i) six efflux transporters, viz., breast cancer resistance protein (BCRP/ABCG2), P-glycoprotein (P-gp/MDR1), and multidrug resistance proteins (MRP1, MRP2, MRP3, and MRP4), and (ii) seven influx transporters, viz., organic cation transporter (OCT1/SO22A1), peptide transporter (PEPT1/SO15A1), apical sodium-bile acid transporter (ASBT/NTCP2), and organic anion transporting peptides (OATP1A2/SO1A2, OATP1B/SO1B1, OATP1B3/SO1B3, and OATP2B1/SO2B1). Various types of descriptors and machine learning methods (classifiers) were evaluated for the development of robust predictive models. Additionally, ensemble models were developed by bagging of homogeneous classifiers and selective fusion of heterogeneous classifiers. It was observed that the latter approach improves the accuracy of substrate/nonsubstrate prediction for transporters (average correct classification rate of more than 0.80 for external validation). Moreover, structural fragments important in determining the substrate specificity across the various transporters were identified. To demonstrate these fragments on the query molecule, contour maps were generated. The prediction efficacy of the developed models was illustrated by a good correlation between the reported logBB value of a molecule and its predicted substrate propensity for blood-brain barrier transporters. Conclusively, this comprehensive modeling analysis can be efficiently employed for the prediction of membrane transporters of a drug, thereby providing insights into its pharmacological profile.
Collapse
Affiliation(s)
- Naeem Shaikh
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER) , S. A. S. Nagar, Mohali, Punjab-160062, India
| | - Mahesh Sharma
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER) , S. A. S. Nagar, Mohali, Punjab-160062, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER) , S. A. S. Nagar, Mohali, Punjab-160062, India
| |
Collapse
|
16
|
Bosc N, Wroblowski B, Meyer C, Bonnet P. Prediction of Protein Kinase-Ligand Interactions through 2.5D Kinochemometrics. J Chem Inf Model 2017; 57:93-101. [PMID: 27983837 DOI: 10.1021/acs.jcim.6b00520] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
So far, 518 protein kinases have been identified in the human genome. They share a common mechanism of protein phosphorylation and are involved in many critical biological processes of eukaryotic cells. Deregulation of the kinase phosphorylation function induces severe illnesses such as cancer, diabetes, or inflammatory diseases. Many actors in the pharmaceutical domain have made significant efforts to design potent and selective protein kinase inhibitors as new potential drugs. Because the ATP binding site is highly conserved in the protein kinase family, the design of selective inhibitors remains a challenge and has negatively impacted the progression of drug candidates to late-stage clinical development. The work presented here adopts a 2.5D kinochemometrics (KCM) approach, derived from proteochemometrics (PCM), in which protein kinases are depicted by a novel 3D descriptor and the ligands by 2D fingerprints. We demonstrate in two examples that the protein descriptor successfully classified protein kinases based on their group membership and their Asp-Phe-Gly (DFG) conformation. We also compared the performance of our models with those obtained from a full 2D KCM model and QSAR models. In both cases, the internal validation of the models demonstrated good capabilities to distinguish "active" from "inactive" protein kinase-ligand pairs. However, the external validation performed on two independent data sets showed that the two statistical models tended to overestimate the number of "inactive" pairs.
Collapse
Affiliation(s)
- Nicolas Bosc
- Institut de Chimie Organique et Analytique (ICOA), UMR CNRS-Université d'Orléans 7311 , Université d'Orléans BP 6759, 45067 Orléans Cedex 2, France
| | - Berthold Wroblowski
- Janssen Research & Development, Janssen Pharmaceutica N.V. , Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Christophe Meyer
- Centre de Recherche Janssen-Cilag , Campus de Maigremont - CS 10615, 27106 Val de Reuil CEDEX, France
| | - Pascal Bonnet
- Institut de Chimie Organique et Analytique (ICOA), UMR CNRS-Université d'Orléans 7311 , Université d'Orléans BP 6759, 45067 Orléans Cedex 2, France
| |
Collapse
|
17
|
Rasti B, Schaduangrat N, Shahangian SS, Nantasenamat C. Exploring the origin of phosphodiesterase inhibition via proteochemometric modeling. RSC Adv 2017. [DOI: 10.1039/c7ra02332d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A proteochemometric study of a set of phosphodiesterase 4B and 4D inhibitors sheds light on the origin of their inhibition and selectivities.
Collapse
Affiliation(s)
- Behnam Rasti
- Department of Microbiology
- Faculty of Basic Sciences
- Lahijan Branch
- Islamic Azad University (IAU)
- Lahijan
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics
- Faculty of Medical Technology
- Mahidol University
- Bangkok 10700
- Thailand
| | - S. Shirin Shahangian
- Department of Biology
- Faculty of Sciences
- University of Guilan
- Rasht 41938-33697
- Iran
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics
- Faculty of Medical Technology
- Mahidol University
- Bangkok 10700
- Thailand
| |
Collapse
|
18
|
Christmann-Franck S, van Westen GJP, Papadatos G, Beltran Escudie F, Roberts A, Overington JP, Domine D. Unprecedently Large-Scale Kinase Inhibitor Set Enabling the Accurate Prediction of Compound-Kinase Activities: A Way toward Selective Promiscuity by Design? J Chem Inf Model 2016; 56:1654-75. [PMID: 27482722 PMCID: PMC5039764 DOI: 10.1021/acs.jcim.6b00122] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Drug discovery programs frequently target members of the human kinome and try to identify small molecule protein kinase inhibitors, primarily for cancer treatment, additional indications being increasingly investigated. One of the challenges is controlling the inhibitors degree of selectivity, assessed by in vitro profiling against panels of protein kinases. We manually extracted, compiled, and standardized such profiles published in the literature: we collected 356 908 data points corresponding to 482 protein kinases, 2106 inhibitors, and 661 patents. We then analyzed this data set in terms of kinome coverage, results reproducibility, popularity, and degree of selectivity of both kinases and inhibitors. We used the data set to create robust proteochemometric models capable of predicting kinase activity (the ligand-target space was modeled with an externally validated RMSE of 0.41 ± 0.02 log units and R02 0.74 ± 0.03), in order to account for missing or unreliable measurements. The influence on the prediction quality of parameters such as number of measurements, Murcko scaffold frequency or inhibitor type was assessed. Interpretation of the models enabled to highlight inhibitors and kinases properties correlated with higher affinities, and an analysis in the context of kinases crystal structures was performed. Overall, the models quality allows the accurate prediction of kinase-inhibitor activities and their structural interpretation, thus paving the way for the rational design of compounds with a targeted selectivity profile.
Collapse
Affiliation(s)
| | - Gerard J P van Westen
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus , Hinxton, Cambridgeshire CB10 1SD, U.K
| | - George Papadatos
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus , Hinxton, Cambridgeshire CB10 1SD, U.K
| | | | | | - John P Overington
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus , Hinxton, Cambridgeshire CB10 1SD, U.K
| | - Daniel Domine
- Merck Serono , Chemin des Mines 9, 1202 Genève, Switzerland
| |
Collapse
|
19
|
Qiu T, Qiu J, Feng J, Wu D, Yang Y, Tang K, Cao Z, Zhu R. The recent progress in proteochemometric modelling: focusing on target descriptors, cross-term descriptors and application scope. Brief Bioinform 2016; 18:125-136. [PMID: 26873661 DOI: 10.1093/bib/bbw004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/09/2015] [Indexed: 12/17/2022] Open
Abstract
As an extension of the conventional quantitative structure activity relationship models, proteochemometric (PCM) modelling is a computational method that can predict the bioactivity relations between multiple ligands and multiple targets. Traditional PCM modelling includes three essential elements: descriptors (including target descriptors, ligand descriptors and cross-term descriptors), bioactivity data and appropriate learning functions that link the descriptors to the bioactivity data. Since its appearance, PCM modelling has developed rapidly over the past decade by taking advantage of the progress of different descriptors and machine learning techniques, along with the increasing amounts of available bioactivity data. Specifically, the new emerging target descriptors and cross-term descriptors not only significantly increased the performance of PCM modelling but also expanded its application scope from traditional protein-ligand interaction to more abundant interactions, including protein-peptide, protein-DNA and even protein-protein interactions. In this review, target descriptors and cross-term descriptors, as well as the corresponding application scope, are intensively summarized. Additionally, we look forward to seeing PCM modelling extend into new application scopes, such as Target-Catalyst-Ligand systems, with the further development of descriptors, machine learning techniques and increasing amounts of available bioactivity data.
Collapse
|
20
|
Volkamer A, Eid S, Turk S, Rippmann F, Fulle S. Identification and Visualization of Kinase-Specific Subpockets. J Chem Inf Model 2016; 56:335-46. [PMID: 26735903 DOI: 10.1021/acs.jcim.5b00627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The identification and design of selective compounds is important for the reduction of unwanted side effects as well as for the development of tool compounds for target validation studies. This is, in particular, true for therapeutically important protein families that possess conserved folds and have numerous members such as kinases. To support the design of selective kinase inhibitors, we developed a novel approach that allows identification of specificity determining subpockets between closely related kinases solely based on their three-dimensional structures. To account for the intrinsic flexibility of the proteins, multiple X-ray structures of the target protein of interest as well as of unwanted off-target(s) are taken into account. The binding pockets of these protein structures are calculated and fused to a combined target and off-target pocket, respectively. Subsequently, shape differences between these two combined pockets are identified via fusion rules. The approach provides a user-friendly visualization of target-specific areas in a binding pocket which should be explored when designing selective compounds. Furthermore, the approach can be easily combined with in silico alanine mutation studies to identify selectivity determining residues. The potential impact of the approach is demonstrated in four retrospective experiments on closely related kinases, i.e., p38α vs Erk2, PAK1 vs PAK4, ITK vs AurA, and BRAF vs VEGFR2. Overall, the presented approach does not require any profiling data for training purposes, provides an intuitive visualization of a large number of protein structures at once, and could also be applied to other target classes.
Collapse
Affiliation(s)
- Andrea Volkamer
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Sameh Eid
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Samo Turk
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Friedrich Rippmann
- Global Computational Chemistry, Merck KGaA , Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Simone Fulle
- BioMed X Innovation Center , Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| |
Collapse
|
21
|
Subramanian V, Prusis P, Xhaard H, Wohlfahrt G. Predictive proteochemometric models for kinases derived from 3D protein field-based descriptors. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00556f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Proteochemometric models of kinases derived from protein fields and ligand 4-point pharmacophoric fingerprints are predictive and visually interpretable.
Collapse
Affiliation(s)
- Vigneshwari Subramanian
- Computer-Aided Drug Design
- Orion Pharma
- FI-02101 Espoo
- Finland
- Division of Pharmaceutical Chemistry and Technology
| | - Peteris Prusis
- Computer-Aided Drug Design
- Orion Pharma
- FI-02101 Espoo
- Finland
| | - Henri Xhaard
- Division of Pharmaceutical Chemistry and Technology
- Faculty of Pharmacy
- University of Helsinki
- FI-00014 Helsinki
- Finland
| | - Gerd Wohlfahrt
- Computer-Aided Drug Design
- Orion Pharma
- FI-02101 Espoo
- Finland
| |
Collapse
|
22
|
Ain QU, Méndez-Lucio O, Ciriano IC, Malliavin T, van Westen GJP, Bender A. Modelling ligand selectivity of serine proteases using integrative proteochemometric approaches improves model performance and allows the multi-target dependent interpretation of features. Integr Biol (Camb) 2015; 6:1023-33. [PMID: 25255469 DOI: 10.1039/c4ib00175c] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Serine proteases, implicated in important physiological functions, have a high intra-family similarity, which leads to unwanted off-target effects of inhibitors with insufficient selectivity. However, the availability of sequence and structure data has now made it possible to develop approaches to design pharmacological agents that can discriminate successfully between their related binding sites. In this study, we have quantified the relationship between 12,625 distinct protease inhibitors and their bioactivity against 67 targets of the serine protease family (20,213 data points) in an integrative manner, using proteochemometric modelling (PCM). The benchmarking of 21 different target descriptors motivated the usage of specific binding pocket amino acid descriptors, which helped in the identification of active site residues and selective compound chemotypes affecting compound affinity and selectivity. PCM models performed better than alternative approaches (models trained using exclusively compound descriptors on all available data, QSAR) employed for comparison with R(2)/RMSE values of 0.64 ± 0.23/0.66 ± 0.20 vs. 0.35 ± 0.27/1.05 ± 0.27 log units, respectively. Moreover, the interpretation of the PCM model singled out various chemical substructures responsible for bioactivity and selectivity towards particular proteases (thrombin, trypsin and coagulation factor 10) in agreement with the literature. For instance, absence of a tertiary sulphonamide was identified to be responsible for decreased selective activity (by on average 0.27 ± 0.65 pChEMBL units) on FA10. Among the binding pocket residues, the amino acids (arginine, leucine and tyrosine) at positions 35, 39, 60, 93, 140 and 207 were observed as key contributing residues for selective affinity on these three targets.
Collapse
Affiliation(s)
- Qurrat U Ain
- Centre for Molecular Informatics, Department of Chemistry, Lensfield Road, CB2 1EW, University of Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
23
|
Assessing protein kinase target similarity: Comparing sequence, structure, and cheminformatics approaches. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1605-16. [PMID: 26001898 DOI: 10.1016/j.bbapap.2015.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 05/08/2015] [Accepted: 05/11/2015] [Indexed: 11/22/2022]
Abstract
In just over two decades, structure based protein kinase inhibitor discovery has grown from trial and error approaches, using individual target structures, to structure and data driven approaches that may aim to optimize inhibition properties across several targets. This is increasingly enabled by the growing availability of potent compounds and kinome-wide binding data. Assessing the prospects for adapting known compounds to new therapeutic uses is thus a key priority for current drug discovery efforts. Tools that can successfully link the diverse information regarding target sequence, structure, and ligand binding properties now accompany a transformation of protein kinase inhibitor research, away from single, block-buster drug models, and toward "personalized medicine" with niche applications and highly specialized research groups. Major hurdles for the transformation to data driven drug discovery include mismatches in data types, and disparities of methods and molecules used; at the core remains the problem that ligand binding energies cannot be predicted precisely from individual structures. However, there is a growing body of experimental data for increasingly successful focussing of efforts: focussed chemical libraries, drug repurposing, polypharmacological design, to name a few. Protein kinase target similarity is easily quantified by sequence, and its relevance to ligand design includes broad classification by key binding sites, evaluation of resistance mutations, and the use of surrogate proteins. Although structural evaluation offers more information, the flexibility of protein kinases, and differences between the crystal and physiological environments may make the use of crystal structures misleading when structures are considered individually. Cheminformatics may enable the "calibration" of sequence and crystal structure information, with statistical methods able to identify key correlates to activity but also here, "the devil is in the details." Examples from specific repurposing and polypharmacology applications illustrate these points. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.
Collapse
|
24
|
Cortés-Ciriano I, Ain QU, Subramanian V, Lenselink EB, Méndez-Lucio O, IJzerman AP, Wohlfahrt G, Prusis P, Malliavin TE, van Westen GJP, Bender A. Polypharmacology modelling using proteochemometrics (PCM): recent methodological developments, applications to target families, and future prospects. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00216d] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Proteochemometric (PCM) modelling is a computational method to model the bioactivity of multiple ligands against multiple related protein targets simultaneously.
Collapse
Affiliation(s)
- Isidro Cortés-Ciriano
- Unité de Bioinformatique Structurale
- Institut Pasteur and CNRS UMR 3825
- Structural Biology and Chemistry Department
- 75 724 Paris
- France
| | - Qurrat Ul Ain
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| | | | - Eelke B. Lenselink
- Division of Medicinal Chemistry
- Leiden Academic Centre for Drug Research
- Leiden
- The Netherlands
| | - Oscar Méndez-Lucio
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry
- Leiden Academic Centre for Drug Research
- Leiden
- The Netherlands
| | - Gerd Wohlfahrt
- Computer-Aided Drug Design
- Orion Pharma
- FIN-02101 Espoo
- Finland
| | - Peteris Prusis
- Computer-Aided Drug Design
- Orion Pharma
- FIN-02101 Espoo
- Finland
| | - Thérèse E. Malliavin
- Unité de Bioinformatique Structurale
- Institut Pasteur and CNRS UMR 3825
- Structural Biology and Chemistry Department
- 75 724 Paris
- France
| | - Gerard J. P. van Westen
- European Molecular Biology Laboratory
- European Bioinformatics Institute
- Wellcome Trust Genome Campus
- Hinxton
- UK
| | - Andreas Bender
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| |
Collapse
|