Automated discovery of GPCR bioactive ligands
Tài liệu tham khảo
Hauser, 2017, Trends in GPCR drug discovery: new agents, targets and indications, Nat Rev Drug Discov, 16, 829, 10.1038/nrd.2017.178
Garland, 2013, Are GPCRs still a source of new targets?, J Biomol Screen, 18, 947, 10.1177/1087057113498418
Thomsen, 2005, Functional assays for screening GPCR targets, Curr Opin Biotechnol, 16, 655
Bjarnadóttir, 2006, Comprehensive repertoire and phylogenetic analysis of the G protein-coupled receptors in human and mouse, Genomics, 88, 263, 10.1016/j.ygeno.2006.04.001
Davenport, 2013, International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: recommendations for new pairings with cognate ligands, Pharmacol Rev, 65, 967, 10.1124/pr.112.007179
Southan, 2016, The IUPHAR/BPS guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands, Nucleic Acids Res, 44, D1054, 10.1093/nar/gkv1037
Sterling, 2015, ZINC 15–ligand discovery for everyone, J Chem Inf Model, 55, 2324, 10.1021/acs.jcim.5b00559
Sunseri, 2016, Pharmit: interactive exploration of chemical space, Nucleic Acids Res, 44, W442, 10.1093/nar/gkw287
Bento, 2014, The ChEMBL bioactivity database: an update, Nucleic Acids Res, 42, D1083, 10.1093/nar/gkt1031
Lipinski, 2000, Drug-like properties and the causes of poor solubility and poor permeability, J Pharmacol Toxicol Methods, 44, 235, 10.1016/S1056-8719(00)00107-6
Baell, 2014, Chemistry: chemical con artists foil drug discovery, Nat News, 513, 481, 10.1038/513481a
Van De Waterbeemd, 2003, ADMET in silico modelling: towards prediction paradise?, Nat Rev Drug Discov, 2, 192, 10.1038/nrd1032
Liu, 2014, Femtosecond crystallography of membrane proteins in the lipidic cubic phase, Philos Trans R Soc B, 369, 10.1098/rstb.2013.0314
Renaud, 2018, Cryo-EM in drug discovery: achievements, limitations and prospects, Nat Rev Drug Discov, 17, 471, 10.1038/nrd.2018.77
Basith, 2018, Exploring G protein-coupled receptors (GPCRs) ligand space via cheminformatics approaches: impact on rational drug design, Front Pharmacol, 9, 1, 10.3389/fphar.2018.00128
Wheatley, 2012, Lifting the lid on GPCRs: the role of extracellular loops: GPCR extracellular loops, Br J Pharmacol, 165, 1688, 10.1111/j.1476-5381.2011.01629.x
Raschka, 2018, Enabling the hypothesis-driven prioritization of ligand candidates in big databases: Screenlamp and its application to GPCR inhibitor discovery for invasive species control, J Comput Aided Mol Des, 32, 415, 10.1007/s10822-018-0100-7
Raschka, 2017
Raschka, 2018, Automated inference of chemical discriminants of biological activity, 307
Liang, 2018, Dominant negative G proteins enhance formation and purification of agonist-GPCR-G protein complexes for structure determination, ACS Pharmacol Transl Sci, 1, 12, 10.1021/acsptsci.8b00017
Klabunde, 2002, Drug design strategies for targeting G-protein-coupled receptors, ChemBioChem, 3, 928, 10.1002/1439-7633(20021004)3:10<928::AID-CBIC928>3.0.CO;2-5
Flohr, 2002, Identification of nonpeptidic urotensin II receptor antagonists by virtual screening based on a pharmacophore model derived from structure–activity relationships and nuclear magnetic resonance studies on urotensin II, J Med Chem, 45, 1799, 10.1021/jm0111043
Evers, 2005, Virtual screening of biogenic amine-binding G-protein coupled receptors: comparative evaluation of protein-and ligand-based virtual screening protocols, J Med Chem, 48, 5448, 10.1021/jm050090o
Bender, 2004, Molecular similarity: a key technique in molecular informatics, Org Biomol Chem, 2, 3204, 10.1039/b409813g
Bender, 2009, How similar are similarity searching methods? A principal component analysis of molecular descriptor space, J Chem Inf Model, 49, 108, 10.1021/ci800249s
Bajusz, 2015, Why is Tanimoto index an appropriate choice for fingerprint-based similarity calculations?, J Cheminform, 7, 20, 10.1186/s13321-015-0069-3
Duan, 2010, Analysis and comparison of 2D fingerprints: insights into database screening performance using eight fingerprint methods, J Mol Graph Model, 29, 157, 10.1016/j.jmgm.2010.05.008
Willett, 2006, Similarity-based virtual screening using 2D fingerprints, Drug Discov Today, 11, 1046, 10.1016/j.drudis.2006.10.005
Awale, 2015, Stereoselective virtual screening of the ZINC database using atom pair 3D-fingerprints, J Cheminform, 7, 3, 10.1186/s13321-014-0051-5
Hawkins, 2007, Comparison of shape-matching and docking as virtual screening tools, J Med Chem, 50, 74, 10.1021/jm0603365
Cortés-Ciriano, 2018, Discovering highly potent molecules from an initial set of inactives using iterative screening, J Chem Inf Model, 58, 2000, 10.1021/acs.jcim.8b00376
Ngo, 2016, Identifying ligands at orphan GPCRs: current status using structure-based approaches: approaches for identifying orphan GPCR ligands, Br J Pharmacol, 173, 2934, 10.1111/bph.13452
Ciancetta, 2016, New trends in inspecting GPCR-ligand recognition process: the contribution of the molecular modeling section (MMS) at the University of Padova, Mol Inform, 35, 440, 10.1002/minf.201501011
Sengupta, 2017, Characterizing clinically relevant natural variants of GPCRs using computational approaches, 187, 10.1016/bs.mcb.2017.07.013
Hadianawala, 2018, Molecular docking, molecular modeling, and molecular dynamics studies of azaisoflavone as dual COX-2 inhibitors and TP receptor antagonists, J Mol Model, 24, 69, 10.1007/s00894-018-3620-0
Dacanay, 2017, Molecular affinity of mabolo extracts to an octopamine receptor of a fruit fly, Molecules, 22, 1677, 10.3390/molecules22101677
Raschka, 2016, Detecting the native ligand orientation by interfacial rigidity: SiteInterlock, Proteins, 84, 1888, 10.1002/prot.25172
Schneider, 2018, Predicting ligand binding poses for low-resolution membrane protein models: perspectives from multiscale simulations, Biochem Biophys Res Commun, 498, 366, 10.1016/j.bbrc.2018.01.160
Roth, 2017, Discovery of new GPCR ligands to illuminate new biology, Nat Chem Biol, 13, 1143, 10.1038/nchembio.2490
Jakubík, 2015, Towards predictive docking at aminergic G-protein coupled receptors, J Mol Model, 21, 284, 10.1007/s00894-015-2824-9
Chen, 2018, The rise of deep learning in drug discovery, Drug Discov Today, 23, 1241, 10.1016/j.drudis.2018.01.039
Ekins, 2014, Bigger data, collaborative tools and the future of predictive drug discovery, J Comput Aided Mol Des, 28, 997, 10.1007/s10822-014-9762-y
Lavecchia, 2015, Machine-learning approaches in drug discovery: methods and applications, Drug Discov Today, 20, 318, 10.1016/j.drudis.2014.10.012
Lima, 2016, Use of machine learning approaches for novel drug discovery, Expert Opin Drug Dis, 11, 225, 10.1517/17460441.2016.1146250
Wale, 2011, Machine learning in drug discovery and development, Drug Dev Res, 72, 112, 10.1002/ddr.20407
Ekins, 2016, The next era: deep learning in pharmaceutical research, Pharm Res, 33, 2594, 10.1007/s11095-016-2029-7
Ching, 2018, Opportunities and obstacles for deep learning in biology and medicine, J R Soc Interface, 15, 10.1098/rsif.2017.0387
Ericksen, 2017, Machine learning consensus scoring improves performance across targets in structure-based virtual screening, J Chem Inf Model, 57, 1579, 10.1021/acs.jcim.7b00153
Wang, 2014, Pairwise input neural network for target-ligand interaction prediction, 67
Raschka, 2018, MLxtend: providing machine learning and data science utilities and extensions to Python’s scientific computing stack, J Open Source Softw, 3, 638, 10.21105/joss.00638
Ribeiro, 2016, Why should I trust you?, 1135
Jacobs, 2001, Protein flexibility predictions using graph theory, Proteins, 44, 150, 10.1002/prot.1081
Bemister-Buffington J, Wolf A, Raschka S, Kuhn LA: Predicting GPCR active and inactive states by rigidity analysis and flexibility profiles. Manuscript in preparation.
Ghasemi, 2018, Neural network and deep-learning algorithms used in QSAR studies: merits and drawbacks, Drug Discov Today, 23, 1784, 10.1016/j.drudis.2018.06.016
Ma, 2015, Deep neural nets as a method for quantitative structure–activity relationships, J Chem Inf Model, 55, 263, 10.1021/ci500747n
Koutsoukas, 2017, Deep-learning: investigating deep neural networks hyper-parameters and comparison of performance to shallow methods for modeling bioactivity data, J Cheminform, 9, 42, 10.1186/s13321-017-0226-y
Lewis, 1998, Naive (Bayes) at forty: the independence assumption in information retrieval, 4
Wang, 2016, ADMET evaluation in drug discovery. 16. Predicting hERG blockers by combining multiple pharmacophores and machine learning approaches, Mol Pharm, 13, 2855, 10.1021/acs.molpharmaceut.6b00471
Nidhi, 2006, Prediction of biological targets for compounds using multiple-category Bayesian models trained on chemogenomics databases, J Chem Inf Model, 46, 1124, 10.1021/ci060003g
Bender, 2007, Analysis of pharmacology data and the prediction of adverse drug reactions and off-target effects from chemical structure, Chem Med Chem, 2, 861, 10.1002/cmdc.200700026
Clark, 2015, Open source Bayesian models. 2. Mining a “big dataset” to create and validate models with ChEMBL, J Chem Inf Model, 55, 1246, 10.1021/acs.jcim.5b00144
Hosmer, 2013
Caster, 2010, Large-scale regression-based pattern discovery: the example of screening the WHO global drug safety database, Stat Anal Data Min, 3, 197, 10.1002/sam.10078
Harpaz, 2012, Novel data-mining methodologies for adverse drug event discovery and analysis, J Clin Pharm Ther, 91, 1010, 10.1038/clpt.2012.50
Kim, 2013, Predicting drug–target interactions using drug–drug interactions, PLoS One, 8
Cristianini, 2000
Kriegl, 2005, A support vector machine approach to classify human cytochrome P450 3A4 inhibitors, J Comput Aided Mol Des, 19, 189, 10.1007/s10822-005-3785-3
Breiman, 2001, Random forests, Mach Learn, 45, 5, 10.1023/A:1010933404324
Svetnik, 2003, Random forest: a classification and regression tool for compound classification and QSAR modeling, J Chem Inf Model, 43, 1947
Svetnik, 2004, Application of Breiman’s random forest to modeling structure–activity relationships of pharmaceutical molecules, 334
Rakers, 2017, Small random forest models for effective chemogenomic active learning, J Med Chem, 18, 124
Korotcov, 2017, Comparison of deep learning with multiple machine learning methods and metrics using diverse drug discovery data sets, Mol Pharm, 14, 4462, 10.1021/acs.molpharmaceut.7b00578
Lenselink, 2017, Beyond the hype: deep neural networks outperform established methods using a ChEMBL bioactivity benchmark set, J Cheminform, 9, 45, 10.1186/s13321-017-0232-0
Mayr, 2018, Large-scale comparison of machine learning methods for drug target prediction on ChEMBL, Chem Sci, 9, 5441, 10.1039/C8SC00148K
Russo, 2018, Comparing multiple machine learning algorithms and metrics for estrogen receptor binding prediction, Mol Pharm, 15, 4361, 10.1021/acs.molpharmaceut.8b00546
Liu, 2018, Practical model selection for prospective virtual screening, J Chem Inf Model, 59, 282, 10.1021/acs.jcim.8b00363
Duvenaud, 2015, Convolutional networks on graphs for learning molecular fingerprints
Hop, 2018, Geometric deep learning autonomously learns chemical features that outperform those engineered by domain experts, Mol Pharm, 15, 4371, 10.1021/acs.molpharmaceut.7b01144
Zhavoronkov, 2018, Artificial intelligence for drug discovery, biomarker development, and generation of novel chemistry, Mol Pharm, 15, 4311, 10.1021/acs.molpharmaceut.8b00930
Raschka, 2017, BioPandas: working with molecular structures in pandas DataFrames, J Open Source Softw, 2, 279, 10.21105/joss.00279
Ochoa, 2013, myChEMBL: a virtual machine implementation of open data and cheminformatics tools, Bioinformatics, 30, 298, 10.1093/bioinformatics/btt666
Cock, 2009, Biopython: freely available Python tools for computational molecular biology and bioinformatics, Bioinformatics, 25, 1422, 10.1093/bioinformatics/btp163
Pedregosa, 2011, Scikit-learn: machine learning in Python, J Mach Learn Res, 12, 2825
Abadi, 2016, TensorFlow: a system for large-scale machine learning, 265
Paszke, 2017, Automatic differentiation in PyTorch, 4
Chuang, 2018, Adversarial controls for scientific machine learning, ACS Chem Biol, 13, 2819, 10.1021/acschembio.8b00881
Raschka, 2018, Protein–ligand interfaces are polarized: discovery of a strong trend for intermolecular hydrogen bonds to favor donors on the protein side with implications for predicting and designing ligand complexes, J Comput Aided Mol Des, 32, 511, 10.1007/s10822-018-0105-2
Feinberg, 2018