Applications of machine learning in drug discovery and development

[1]  A. Butte,et al.  A call for deep-learning healthcare , 2019, Nature Medicine.

[2]  J. Dean,et al.  A guide to deep learning in healthcare , 2019, Nature Medicine.

[3]  Volkan Atalay,et al.  Recent applications of deep learning and machine intelligence on in silico drug discovery: methods, tools and databases , 2018, Briefings Bioinform..

[4]  Chi Heem Wong,et al.  Estimation of clinical trial success rates and related parameters , 2018, Biostatistics.

[5]  Olexandr Isayev,et al.  Transforming Computational Drug Discovery with Machine Learning and AI. , 2018, ACS medicinal chemistry letters.

[6]  Jin Gu,et al.  VASC: Dimension Reduction and Visualization of Single-cell RNA-seq Data by Deep Variational Autoencoder , 2018, Genom. Proteom. Bioinform..

[7]  Geoffrey E. Hinton Deep Learning-A Technology With the Potential to Transform Health Care. , 2018, JAMA.

[8]  N. Razavian,et al.  Classification and mutation prediction from non–small cell lung cancer histopathology images using deep learning , 2018, Nature Medicine.

[9]  A. Madabhushi,et al.  Spatial Architecture and Arrangement of Tumor-Infiltrating Lymphocytes for Predicting Likelihood of Recurrence in Early-Stage Non–Small Cell Lung Cancer , 2018, Clinical Cancer Research.

[10]  Mary E. Haas,et al.  Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations , 2018, Nature Genetics.

[11]  R. Morita,et al.  Multi-omics monitoring of drug response in rheumatoid arthritis in pursuit of molecular remission , 2018, Nature Communications.

[12]  A. Zhavoronkov,et al.  Machine Learning on Human Muscle Transcriptomic Data for Biomarker Discovery and Tissue-Specific Drug Target Identification , 2018, Front. Genet..

[13]  A. Madabhushi,et al.  Nuclear shape and orientation features from H&E images predict survival in early-stage estrogen receptor-positive breast cancers , 2018, Laboratory Investigation.

[14]  Mary E. Haas,et al.  Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations , 2018, Nature Genetics.

[15]  Hugo Ceulemans,et al.  Large-scale comparison of machine learning methods for drug target prediction on ChEMBL† †Electronic supplementary information (ESI) available: Overview, Data Collection and Clustering, Methods, Results, Appendix. See DOI: 10.1039/c8sc00148k , 2018, Chemical science.

[16]  Virginia B. Kraus,et al.  Biomarkers as drug development tools: discovery, validation, qualification and use , 2018, Nature Reviews Rheumatology.

[17]  Thomas Blaschke,et al.  The rise of deep learning in drug discovery. , 2018, Drug discovery today.

[18]  Colin C. Pritchard,et al.  Significance and implications of FDA approval of pembrolizumab for biomarker-defined disease , 2018, Journal of Immunotherapy for Cancer.

[19]  Michael Peyton,et al.  Chemistry-First Approach for Nomination of Personalized Treatment in Lung Cancer , 2018, Cell.

[20]  Yongfeng Huang,et al.  Clinical Assistant Diagnosis for Electronic Medical Record Based on Convolutional Neural Network , 2018, Scientific Reports.

[21]  Rajarsi R. Gupta,et al.  Spatial Organization and Molecular Correlation of Tumor-Infiltrating Lymphocytes Using Deep Learning on Pathology Images. , 2018, Cell reports.

[22]  Andrew Janowczyk,et al.  A deep-learning classifier identifies patients with clinical heart failure using whole-slide images of H&E tissue , 2018, PloS one.

[23]  Mike Preuss,et al.  Planning chemical syntheses with deep neural networks and symbolic AI , 2017, Nature.

[24]  Sepp Hochreiter,et al.  Fréchet ChemNet Distance: A Metric for Generative Models for Molecules in Drug Discovery , 2018, J. Chem. Inf. Model..

[25]  Seth A Ament,et al.  Transcriptional regulatory networks underlying gene expression changes in Huntington's disease , 2018, Molecular systems biology.

[26]  Dmitrii Bychkov,et al.  Deep learning based tissue analysis predicts outcome in colorectal cancer , 2018, Scientific Reports.

[27]  M. Hutson Artificial intelligence faces reproducibility crisis. , 2018, Science.

[28]  Amir Barati Farimani,et al.  Binding Pathway of Opiates to μ-Opioid Receptors Revealed by Machine Learning , 2018, 1804.08206.

[29]  Harry Hemingway,et al.  Machine learning models in electronic health records can outperform conventional survival models for predicting patient mortality in coronary artery disease , 2018, bioRxiv.

[30]  David J. Nicholls,et al.  Impact of a five-dimensional framework on R&D productivity at AstraZeneca , 2018, Nature Reviews Drug Discovery.

[31]  Thomas George Olsen,et al.  Diagnostic Performance of Deep Learning Algorithms Applied to Three Common Diagnoses in Dermatopathology , 2018, Journal of pathology informatics.

[32]  Bo Zhu,et al.  MR fingerprinting Deep RecOnstruction NEtwork (DRONE) , 2017, Magnetic resonance in medicine.

[33]  Joseph Gomes,et al.  MoleculeNet: a benchmark for molecular machine learning† †Electronic supplementary information (ESI) available. See DOI: 10.1039/c7sc02664a , 2017, Chemical science.

[34]  Pankaj Agarwal,et al.  Systematic interrogation of diverse Omic data reveals interpretable, robust, and generalizable transcriptomic features of clinically successful therapeutic targets , 2017, bioRxiv.

[35]  Russ P Carstens,et al.  ESRP1 Mutations Cause Hearing Loss due to Defects in Alternative Splicing that Disrupt Cochlear Development. , 2017, Developmental cell.

[36]  Olaf Hellwich,et al.  Deep convolutional neural networks for automatic classification of gastric carcinoma using whole slide images in digital histopathology , 2017, Comput. Medical Imaging Graph..

[37]  John F. McDonald,et al.  Open source machine-learning algorithms for the prediction of optimal cancer drug therapies , 2017, PloS one.

[38]  Ziv Bar-Joseph,et al.  Dhaka: Variational Autoencoder for Unmasking Tumor Heterogeneity from Single Cell Genomic Data , 2017, bioRxiv.

[39]  Taner Z. Sen,et al.  The art of curation at a biological database: Principles and application , 2017 .

[40]  A. Condon,et al.  Interpretable dimensionality reduction of single cell transcriptome data with deep generative models , 2017, bioRxiv.

[41]  P. Sanseau,et al.  In silico prediction of novel therapeutic targets using gene–disease association data , 2017, Journal of Translational Medicine.

[42]  Casey S. Greene,et al.  Extracting a Biologically Relevant Latent Space from Cancer Transcriptomes with Variational Autoencoders , 2017, bioRxiv.

[43]  Sergey Nikolenko,et al.  druGAN: An Advanced Generative Adversarial Autoencoder Model for de Novo Generation of New Molecules with Desired Molecular Properties in Silico. , 2017, Molecular pharmaceutics.

[44]  A. Madabhushi,et al.  An oral cavity squamous cell carcinoma quantitative histomorphometric-based image classifier of nuclear morphology can risk stratify patients for disease-specific survival , 2017, Modern Pathology.

[45]  Andreas Verras,et al.  Is Multitask Deep Learning Practical for Pharma? , 2017, J. Chem. Inf. Model..

[46]  George Papadatos,et al.  Beyond the hype: deep neural networks outperform established methods using a ChEMBL bioactivity benchmark set , 2017, bioRxiv.

[47]  Phillip G. Montgomery,et al.  Defining a Cancer Dependency Map , 2017, Cell.

[48]  P. Sanseau,et al.  In silico prediction of novel therapeutic targets using gene–disease association data , 2017, Journal of Translational Medicine.

[49]  Haiying Xu,et al.  Abstract 662: The differential association of PD-1, PD-L1, and CD8+ cells with response to pembrolizumab and presence of Merkel cell polyomavirus (MCPyV) in patients with Merkel cell carcinoma (MCC) , 2017 .

[50]  Xian Zhang,et al.  A multi‐scale convolutional neural network for phenotyping high‐content cellular images , 2017, Bioinform..

[51]  Andrew Janowczyk,et al.  A deep learning based strategy for identifying and associating mitotic activity with gene expression derived risk categories in estrogen receptor positive breast cancers , 2017, Cytometry. Part A : the journal of the International Society for Analytical Cytology.

[52]  Andrew H. Beck,et al.  Morphoproteomic Characterization of Lung Squamous Cell Carcinoma Fragmentation, a Histological Marker of Increased Tumor Invasiveness. , 2017, Cancer research.

[53]  D. Mohr,et al.  Personal Sensing: Understanding Mental Health Using Ubiquitous Sensors and Machine Learning. , 2017, Annual review of clinical psychology.

[54]  Ranadip Pal,et al.  IntegratedMRF: random forest‐based framework for integrating prediction from different data types , 2017, Bioinform..

[55]  Qi Wang,et al.  A novel framework for the identification of drug target proteins: Combining stacked auto-encoders with a biased support vector machine , 2017, PloS one.

[56]  Barend Mons,et al.  Bridging the translational innovation gap through good biomarker practice , 2017, Nature Reviews Drug Discovery.

[57]  Thomas Blaschke,et al.  Molecular de-novo design through deep reinforcement learning , 2017, Journal of Cheminformatics.

[58]  Anant Madabhushi,et al.  Accurate and reproducible invasive breast cancer detection in whole-slide images: A Deep Learning approach for quantifying tumor extent , 2017, Scientific Reports.

[59]  R. Dobson,et al.  Characterisation of mental health conditions in social media using Informed Deep Learning , 2017, Scientific Reports.

[60]  Wei Q. Deng,et al.  A machine-learning heuristic to improve gene score prediction of polygenic traits , 2017, Scientific Reports.

[61]  Jun S. Song,et al.  Maximum entropy methods for extracting the learned features of deep neural networks , 2017, bioRxiv.

[62]  Feng Lin,et al.  Low-Dose CT With a Residual Encoder-Decoder Convolutional Neural Network , 2017, IEEE Transactions on Medical Imaging.

[63]  Yoseph Barash,et al.  Integrative deep models for alternative splicing , 2017, bioRxiv.

[64]  Hyunju Lee,et al.  An analysis of disease-gene relationship from Medline abstracts by DigSee , 2017, Scientific Reports.

[65]  Saeed Hassanpour,et al.  Deep Learning for Classification of Colorectal Polyps on Whole-slide Images , 2017, Journal of pathology informatics.

[66]  Gautier Koscielny,et al.  Open Targets: a platform for therapeutic target identification and validation , 2016, Nucleic Acids Res..

[67]  George Papadatos,et al.  The ChEMBL database in 2017 , 2016, Nucleic Acids Res..

[68]  George Lee,et al.  Nuclear Shape and Architecture in Benign Fields Predict Biochemical Recurrence in Prostate Cancer Patients Following Radical Prostatectomy: Preliminary Findings. , 2016, European urology focus.

[69]  Bo Wang,et al.  Visualization and analysis of single-cell RNA-seq data by kernel-based similarity learning , 2016, Nature Methods.

[70]  Mannudeep K. Kalra,et al.  Low-Dose CT with a Residual Encoder-Decoder Convolutional Neural Network (RED-CNN) , 2017, ArXiv.

[71]  Yasen Jiao,et al.  Performance measures in evaluating machine learning based bioinformatics predictors for classifications , 2016, Quantitative Biology.

[72]  Pankaj Agarwal,et al.  Systematic Analysis of Drug Targets Confirms Expression in Disease-Relevant Tissues , 2016, Scientific Reports.

[73]  Stefan Kramer,et al.  Filtered circular fingerprints improve either prediction or runtime performance while retaining interpretability , 2016, Journal of Cheminformatics.

[74]  L. Pusztai,et al.  Quantitative assessment of the spatial heterogeneity of tumor-infiltrating lymphocytes in breast cancer , 2016, Breast Cancer Research.

[75]  Emanuel J. V. Gonçalves,et al.  A Landscape of Pharmacogenomic Interactions in Cancer , 2016, Cell.

[76]  O. Stegle,et al.  Deep learning for computational biology , 2016, Molecular systems biology.

[77]  Andrew Janowczyk,et al.  Automated Tubule Nuclei Quantification and Correlation with Oncotype DX risk categories in ER+ Breast Cancer Whole Slide Images , 2016, Scientific Reports.

[78]  Juan González-Vallinas,et al.  A new view of transcriptome complexity and regulation through the lens of local splicing variations , 2016, eLife.

[79]  C. Greene,et al.  ADAGE-Based Integration of Publicly Available Pseudomonas aeruginosa Gene Expression Data with Denoising Autoencoders Illuminates Microbe-Host Interactions , 2016, mSystems.

[80]  Andrew Janowczyk,et al.  Deep learning for digital pathology image analysis: A comprehensive tutorial with selected use cases , 2016, Journal of pathology informatics.

[81]  N. Linder,et al.  Antibody-supervised deep learning for quantification of tumor-infiltrating immune cells in hematoxylin and eosin stained breast cancer samples , 2016, Journal of pathology informatics.

[82]  Samuel Kaski,et al.  Sparse group factor analysis for biclustering of multiple data sources , 2015, Bioinform..

[83]  Ney Lemke,et al.  Prediction of Druggable Proteins Using Machine Learning and Systems Biology: A Mini-Review , 2015, Front. Physiol..

[84]  David Allman,et al.  Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy. , 2015, Cancer discovery.

[85]  E. Pierson,et al.  ZIFA: Dimensionality reduction for zero-inflated single-cell gene expression analysis , 2015, Genome Biology.

[86]  Mulin Jun Li,et al.  Nature Genetics Advance Online Publication a N a Ly S I S the Support of Human Genetic Evidence for Approved Drug Indications , 2022 .

[87]  Yuri Nikolsky,et al.  Development of a Drug-Response Modeling Framework to Identify Cell Line Derived Translational Biomarkers That Can Predict Treatment Outcome to Erlotinib or Sorafenib , 2015, PloS one.

[88]  Geoffrey E. Hinton,et al.  Deep Learning , 2015, Nature.

[89]  Sepp Hochreiter,et al.  Toxicity Prediction using Deep Learning , 2015, ArXiv.

[90]  Robert P. Sheridan,et al.  Deep Neural Nets as a Method for Quantitative Structure-Activity Relationships , 2015, J. Chem. Inf. Model..

[91]  Vijay S. Pande,et al.  Massively Multitask Networks for Drug Discovery , 2015, ArXiv.

[92]  HA Hejase,et al.  Improving Drug Sensitivity Prediction Using Different Types of Data , 2015, CPT: pharmacometrics & systems pharmacology.

[93]  B. Frey,et al.  The human splicing code reveals new insights into the genetic determinants of disease , 2015, Science.

[94]  Nci Dream Community A community effort to assess and improve drug sensitivity prediction algorithms , 2014 .

[95]  Philip M. Kim,et al.  A systematic approach to identify novel cancer drug targets using machine learning, inhibitor design and high-throughput screening , 2014, Genome Medicine.

[96]  Núria Queralt-Rosinach,et al.  Extraction of relations between genes and diseases from text and large-scale data analysis: implications for translational research , 2014, BMC Bioinformatics.

[97]  Paul Czodrowski,et al.  Count on kappa , 2014, Journal of Computer-Aided Molecular Design.

[98]  Yuan Wang,et al.  Using Information from Historical High-Throughput Screens to Predict Active Compounds , 2014, J. Chem. Inf. Model..

[99]  Brendan J. Frey,et al.  Deep learning of the tissue-regulated splicing code , 2014, Bioinform..

[100]  Nitish Srivastava,et al.  Dropout: a simple way to prevent neural networks from overfitting , 2014, J. Mach. Learn. Res..

[101]  Maqc Consortium The MicroArray Quality Control ( MAQC )-II study of common practices for the development and validation of microarray-based predictive models , 2012 .

[102]  Andrew H. Beck,et al.  Systematic Analysis of Breast Cancer Morphology Uncovers Stromal Features Associated with Survival , 2011, Science Translational Medicine.

[103]  Edward S. Kim,et al.  The BATTLE trial: personalizing therapy for lung cancer. , 2011, Cancer discovery.

[104]  P.A.C.R. Costa,et al.  A machine learning approach for genome-wide prediction of morbid and druggable human genes based on systems-level data , 2010, BMC Genomics.

[105]  Kevin C. Dorff,et al.  The MicroArray Quality Control (MAQC)-II study of common practices for the development and validation of microarray-based predictive models , 2010, Nature Biotechnology.

[106]  S. Koscielny Why Most Gene Expression Signatures of Tumors Have Not Been Useful in the Clinic , 2010, Science Translational Medicine.

[107]  Andrew J. Doig,et al.  Properties and identification of human protein drug targets , 2009, Bioinform..

[108]  Laurence Lodé,et al.  Prediction of survival in multiple myeloma based on gene expression profiles reveals cell cycle and chromosomal instability signatures in high-risk patients and hyperdiploid signatures in low-risk patients: a study of the Intergroupe Francophone du Myélome. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[109]  John D. Shaughnessy,et al.  High-risk myeloma: a gene expression based risk-stratification model for newly diagnosed multiple myeloma treated with high-dose therapy is predictive of outcome in relapsed disease treated with single-agent bortezomib or high-dose dexamethasone. , 2008, Blood.

[110]  Luhua Lai,et al.  Prediction of potential drug targets based on simple sequence properties , 2007, BMC Bioinformatics.

[111]  Michael J. Keiser,et al.  Relating protein pharmacology by ligand chemistry , 2007, Nature Biotechnology.

[112]  Anthony Boral,et al.  Gene expression profiling and correlation with outcome in clinical trials of the proteasome inhibitor bortezomib. , 2006, Blood.

[113]  Yongsheng Huang,et al.  A validated gene expression model of high-risk multiple myeloma is defined by deregulated expression of genes mapping to chromosome 1. , 2006, Blood.

[114]  John Crowley,et al.  The molecular classification of multiple myeloma. , 2006, Blood.

[115]  Geoffrey E. Hinton,et al.  Reducing the Dimensionality of Data with Neural Networks , 2006, Science.

[116]  B. Honig,et al.  On the nature of cavities on protein surfaces: Application to the identification of drug‐binding sites , 2006, Proteins.

[117]  R W Veltri,et al.  Quantitative nuclear grade (QNG): A new image analysis‐based biomarker of clinically relevant nuclear structure alterations , 2000, Journal of cellular biochemistry. Supplement.