Fusing Dual-Event Data Sets for Mycobacterium tuberculosis Machine Learning Models and Their Evaluation
暂无分享,去创建一个
[1] Vinod Scaria,et al. Predictive models for anti-tubercular molecules using machine learning on high-throughput biological screening datasets , 2011, BMC Research Notes.
[2] David Beer,et al. A High-Throughput Screen To Identify Inhibitors of ATP Homeostasis in Non-replicating Mycobacterium tuberculosis , 2012, ACS chemical biology.
[3] Ying Zhang,et al. The magic bullets and tuberculosis drug targets. , 2005, Annual review of pharmacology and toxicology.
[4] Egon L. Willighagen,et al. Towards interoperable and reproducible QSAR analyses: Exchange of datasets , 2010, J. Cheminformatics.
[5] D. Bojanic,et al. Impact of high-throughput screening in biomedical research , 2011, Nature Reviews Drug Discovery.
[6] A. Bender,et al. Analysis of Pharmacology Data and the Prediction of Adverse Drug Reactions and Off‐Target Effects from Chemical Structure , 2007, ChemMedChem.
[7] Eric Arnoult,et al. The challenge of new drug discovery for tuberculosis , 2011, Nature.
[8] I. Orme,et al. Comprehensive analysis of methods used for the evaluation of compounds against Mycobacterium tuberculosis. , 2012, Tuberculosis.
[9] Vinod Scaria,et al. Computational models for in-vitro anti-tubercular activity of molecules based on high-throughput chemical biology screening datasets , 2012, BMC pharmacology.
[10] Pedro M Alzari,et al. Rising standards for tuberculosis drug development. , 2008, Trends in pharmacological sciences.
[11] Lynn Rasmussen,et al. High-throughput screening for inhibitors of Mycobacterium tuberculosis H37Rv. , 2009, Tuberculosis.
[12] Peter Willett,et al. Combination of Similarity Rankings Using Data Fusion , 2013, J. Chem. Inf. Model..
[13] Eszter Hazai,et al. Predicting P-Glycoprotein-Mediated Drug Transport Based On Support Vector Machine and Three-Dimensional Crystal Structure of P-glycoprotein , 2011, PloS one.
[14] Lynn Rasmussen,et al. High throughput screening of a library based on kinase inhibitor scaffolds against Mycobacterium tuberculosis H37Rv. , 2012, Tuberculosis.
[15] Kristin P. Bennett,et al. Support vector machines: hype or hallelujah? , 2000, SKDD.
[16] Sean Ekins,et al. A collaborative database and computational models for tuberculosis drug discovery. , 2010, Molecular bioSystems.
[17] Rieko Arimoto,et al. Development of CYP3A4 Inhibition Models: Comparisons of Machine-Learning Techniques and Molecular Descriptors , 2005, Journal of biomolecular screening.
[18] Sean Ekins,et al. When pharmaceutical companies publish large datasets: an abundance of riches or fool's gold? , 2010, Drug discovery today.
[19] Kathrin Heikamp,et al. Comparison of Confirmed Inactive and Randomly Selected Compounds as Negative Training Examples in Support Vector Machine-Based Virtual Screening , 2013, J. Chem. Inf. Model..
[20] Sean Ekins,et al. Combining Cheminformatics Methods and Pathway Analysis to Identify Molecules with Whole-Cell Activity Against Mycobacterium Tuberculosis , 2012, Pharmaceutical Research.
[21] Christoph Helma,et al. Classification of cytochrome p(450) activities using machine learning methods. , 2009, Molecular pharmaceutics.
[22] Sean Ekins,et al. Computational models for tuberculosis drug discovery. , 2013, Methods in molecular biology.
[23] Ola Spjuth,et al. Open source drug discovery with bioclipse. , 2012, Current topics in medicinal chemistry.
[24] Feixiong Cheng,et al. In silico Prediction of Chemical Ames Mutagenicity , 2012, J. Chem. Inf. Model..
[25] Robert A. Field,et al. New Small-Molecule Synthetic Antimycobacterials , 2005, Antimicrobial Agents and Chemotherapy.
[26] Sean Ekins,et al. Integrated in silico-in vitro strategy for addressing cytochrome P450 3A4 time-dependent inhibition. , 2010, Chemical research in toxicology.
[27] Philip Prathipati,et al. Global Bayesian Models for the Prioritization of Antitubercular Agents , 2008, J. Chem. Inf. Model..
[28] Alexander Tropsha,et al. Chembench: a cheminformatics workbench , 2010, Bioinform..
[29] Damiano Banfi,et al. Leads for antitubercular compounds from kinase inhibitor library screens. , 2010, Tuberculosis.
[30] Ivan Rusyn,et al. Modeling liver-related adverse effects of drugs using knearest neighbor quantitative structure-activity relationship method. , 2010, Chemical research in toxicology.
[31] Sarah R. Langdon,et al. Predicting cytotoxicity from heterogeneous data sources with Bayesian learning , 2010, J. Cheminformatics.
[32] Lynn Rasmussen,et al. Antituberculosis activity of the molecular libraries screening center network library. , 2009, Tuberculosis.
[33] Sean Ekins,et al. Validating New Tuberculosis Computational Models with Public Whole Cell Screening Aerobic Activity Datasets , 2011, Pharmaceutical Research.
[34] D Haussler,et al. Knowledge-based analysis of microarray gene expression data by using support vector machines. , 2000, Proceedings of the National Academy of Sciences of the United States of America.
[35] Raimund Mannhold,et al. QSAR modeling and data mining link Torsades de Pointes risk to the interplay of extent of metabolism, active transport, and HERG liability. , 2012, Molecular pharmaceutics.
[36] Anthony E. Klon,et al. Improved Naïve Bayesian Modeling of Numerical Data for Absorption, Distribution, Metabolism and Excretion (ADME) Property Prediction , 2006, J. Chem. Inf. Model..
[37] Indira Ghosh,et al. Developing an Antituberculosis Compounds Database and Data Mining in the Search of a Motif Responsible for the Activity of a Diverse Class of Antituberculosis Agents , 2006, J. Chem. Inf. Model..
[38] Ramón García-Domenech,et al. Search of Chemical Scaffolds for Novel Antituberculosis Agents , 2005, Journal of biomolecular screening.
[39] Sean Ekins,et al. Computational Models for Neglected Diseases: Gaps and Opportunities , 2013, Pharmaceutical Research.
[40] Hanna Geppert,et al. Current Trends in Ligand-Based Virtual Screening: Molecular Representations, Data Mining Methods, New Application Areas, and Performance Evaluation , 2010, J. Chem. Inf. Model..
[41] Enrique Molina Pérez,et al. Design of novel antituberculosis compounds using graph-theoretical and substructural approaches , 2009, Molecular Diversity.
[42] Bernd Beck,et al. A support vector machine approach to classify human cytochrome P450 3A4 inhibitors , 2005, J. Comput. Aided Mol. Des..
[43] B. Barrell,et al. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence , 1998, Nature.
[44] Marlene T. Kim,et al. Predicting chemical ocular toxicity using a combinatorial QSAR approach. , 2012, Chemical research in toxicology.
[45] Franco Lombardo,et al. A hybrid mixture discriminant analysis-random forest computational model for the prediction of volume of distribution of drugs in human. , 2006, Journal of medicinal chemistry.
[46] D. Pompliano,et al. Drugs for bad bugs: confronting the challenges of antibacterial discovery , 2007, Nature Reviews Drug Discovery.
[47] Barry A. Bunin,et al. Bayesian models leveraging bioactivity and cytotoxicity information for drug discovery. , 2013, Chemistry & biology.
[48] D. Rogers,et al. Using Extended-Connectivity Fingerprints with Laplacian-Modified Bayesian Analysis in High-Throughput Screening Follow-Up , 2005, Journal of biomolecular screening.
[49] Joel S. Freundlich,et al. Computational databases, pathway and cheminformatics tools for tuberculosis drug discovery. , 2011, Trends in Microbiology.
[50] Sean Ekins,et al. Computational mapping tools for drug discovery. , 2009, Drug discovery today.
[51] Hinrich W. H. Göhlmann,et al. A Diarylquinoline Drug Active on the ATP Synthase of Mycobacterium tuberculosis , 2005, Science.
[52] Sean Ekins,et al. Classification of Metabolites with Kernel-Partial Least Squares (K-PLS) , 2007, Drug Metabolism and Disposition.
[53] Dharmaranjan Sriram,et al. Enhanced ranking of PknB Inhibitors using data fusion methods , 2013, Journal of Cheminformatics.
[54] Sean Ekins,et al. Identification and Validation of Novel Human Pregnane X Receptor Activators among Prescribed Drugs via Ligand-Based Virtual Screening , 2011, Drug Metabolism and Disposition.
[55] James R. Brown,et al. Thousands of chemical starting points for antimalarial lead identification , 2010, Nature.
[56] Sean Ekins,et al. Using Open Source Computational Tools for Predicting Human Metabolic Stability and Additional Absorption, Distribution, Metabolism, Excretion, and Toxicity Properties , 2010, Drug Metabolism and Disposition.
[57] Kuo-Chen Chou,et al. Support vector machines for the classification and prediction of β‐turn types , 2002, Journal of peptide science : an official publication of the European Peptide Society.
[58] Sean Ekins,et al. A Predictive Ligand-Based Bayesian Model for Human Drug-Induced Liver Injury , 2010, Drug Metabolism and Disposition.
[59] Sean Ekins,et al. Molecular Determinants of Ligand Selectivity for the Human Multidrug and Toxin Extruder Proteins MATE1 and MATE2-K , 2012, Journal of Pharmacology and Experimental Therapeutics.
[60] Sean Ekins,et al. Novel web-based tools combining chemistry informatics, biology and social networks for drug discovery. , 2009, Drug discovery today.
[61] Sean Ekins,et al. Structure-activity relationship for FDA approved drugs as inhibitors of the human sodium taurocholate cotransporting polypeptide (NTCP). , 2013, Molecular pharmaceutics.
[62] Sean Ekins,et al. The Collaborative Drug Discovery (CDD) database. , 2013, Methods in molecular biology.
[63] Praveen M. Bahadduri,et al. Rapid Identification of P-glycoprotein Substrates and Inhibitors , 2006, Drug Metabolism and Disposition.
[64] T. Alber,et al. Depletion of antibiotic targets has widely varying effects on growth , 2011, Proceedings of the National Academy of Sciences.
[65] Pierre Baldi,et al. A CROC stronger than ROC: measuring, visualizing and optimizing early retrieval , 2010, Bioinform..
[66] David Rogers,et al. Cheminformatics analysis and learning in a data pipelining environment , 2006, Molecular Diversity.
[67] Chin Yee Liew,et al. Mixed learning algorithms and features ensemble in hepatotoxicity prediction , 2011, J. Comput. Aided Mol. Des..
[68] Noriaki Iwase,et al. Identification of novel inhibitors of M. tuberculosis growth using whole cell based high-throughput screening. , 2012, ACS chemical biology.
[69] Barry A. Bunin,et al. Chemical Space: Missing Pieces in Cheminformatics , 2010, Pharmaceutical Research.
[70] Sean Ekins,et al. Computational Approaches That Predict Metabolic Intermediate Complex Formation with CYP3A4 (+b5) , 2007, Drug Metabolism and Disposition.
[71] Klaus-Robert Müller,et al. Benchmark Data Set for in Silico Prediction of Ames Mutagenicity , 2009, J. Chem. Inf. Model..
[72] Marianne Terrot,et al. Combinatorial lead optimization of [1,2]-diamines based on ethambutol as potential antituberculosis preclinical candidates. , 2003, Journal of combinatorial chemistry.
[73] Sean Ekins,et al. Combining Computational Methods for Hit to Lead Optimization in Mycobacterium Tuberculosis Drug Discovery , 2013, Pharmaceutical Research.
[74] Sean Ekins,et al. Analysis and hit filtering of a very large library of compounds screened against Mycobacterium tuberculosis. , 2010, Molecular bioSystems.
[75] Alex M. Clark,et al. TB Mobile: a mobile app for anti-tuberculosis molecules with known targets , 2013, Journal of Cheminformatics.
[76] Bernard F. Buxton,et al. Drug Design by Machine Learning: Support Vector Machines for Pharmaceutical Data Analysis , 2001, Comput. Chem..
[77] Sean Ekins,et al. Enhancing Hit Identification in Mycobacterium tuberculosis Drug Discovery Using Validated Dual-Event Bayesian Models , 2013, PloS one.
[78] Egon L. Willighagen,et al. Bioclipse: an open source workbench for chemo- and bioinformatics , 2007, BMC Bioinformatics.
[79] Alfonso Mendoza,et al. Fueling Open-Source Drug Discovery: 177 Small-Molecule Leads against Tuberculosis , 2013, ChemMedChem.