DeepSynergy: predicting anti-cancer drug synergy with Deep Learning

Abstract Motivation While drug combination therapies are a well-established concept in cancer treatment, identifying novel synergistic combinations is challenging due to the size of combinatorial space. However, computational approaches have emerged as a time- and cost-efficient way to prioritize combinations to test, based on recently available large-scale combination screening data. Recently, Deep Learning has had an impact in many research areas by achieving new state-of-the-art model performance. However, Deep Learning has not yet been applied to drug synergy prediction, which is the approach we present here, termed DeepSynergy. DeepSynergy uses chemical and genomic information as input information, a normalization strategy to account for input data heterogeneity, and conical layers to model drug synergies. Results DeepSynergy was compared to other machine learning methods such as Gradient Boosting Machines, Random Forests, Support Vector Machines and Elastic Nets on the largest publicly available synergy dataset with respect to mean squared error. DeepSynergy significantly outperformed the other methods with an improvement of 7.2% over the second best method at the prediction of novel drug combinations within the space of explored drugs and cell lines. At this task, the mean Pearson correlation coefficient between the measured and the predicted values of DeepSynergy was 0.73. Applying DeepSynergy for classification of these novel drug combinations resulted in a high predictive performance of an AUC of 0.90. Furthermore, we found that all compared methods exhibit low predictive performance when extrapolating to unexplored drugs or cell lines, which we suggest is due to limitations in the size and diversity of the dataset. We envision that DeepSynergy could be a valuable tool for selecting novel synergistic drug combinations. Availability and implementation DeepSynergy is available via www.bioinf.jku.at/software/DeepSynergy. Supplementary information Supplementary data are available at Bioinformatics online.

[1]  Christian Melander,et al.  Combination approaches to combat multidrug-resistant bacteria. , 2013, Trends in biotechnology.

[2]  Knut Baumann,et al.  Reliable estimation of prediction errors for QSAR models under model uncertainty using double cross-validation , 2014, Journal of Cheminformatics.

[3]  César López-Camarillo,et al.  Oncogenomics and Cancer Proteomics - Novel Approaches in Biomarkers Discovery and Therapeutic Targets in Cancer , 2013 .

[4]  S. Ng,et al.  Bexarotene (LGD1069, Targretin), a selective retinoid X receptor agonist, prevents and reverses gemcitabine resistance in NSCLC cells by modulating gene amplification. , 2007, Cancer research.

[5]  B. Al-Lazikani,et al.  Combinatorial drug therapy for cancer in the post-genomic era , 2012, Nature Biotechnology.

[6]  Y. Miura,et al.  Schedule-dependent interaction between paclitaxel and doxorubicin in human cancer cell lines in vitro. , 1995, European journal of cancer.

[7]  H. Zou,et al.  Regularization and variable selection via the elastic net , 2005 .

[8]  Xin Chen,et al.  DCDB 2.0: a major update of the drug combination database , 2014, Database J. Biol. Databases Curation.

[9]  Lawrence A. Donehower,et al.  Combinatorial therapy discovery using mixed integer linear programming , 2014, Bioinform..

[10]  Peter J. Park,et al.  Systematic Identification of Synergistic Drug Pairs Targeting HIV , 2012, Nature Biotechnology.

[11]  Jürgen Bajorath,et al.  Integration of virtual and high-throughput screening , 2002, Nature Reviews Drug Discovery.

[12]  Ting-Chao Chou,et al.  Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies , 2006, Pharmacological Reviews.

[13]  Sam Michael,et al.  High-throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell–like diffuse large B-cell lymphoma cells , 2014, Proceedings of the National Academy of Sciences.

[14]  Krister Wennerberg,et al.  Corrigendum to “Searching for drug synergy in complex dose–response landscapes using an interaction potency model” [Comput. Struct. Biotechnol. J. 13 (2015) 504–513] , 2017, Computational and structural biotechnology journal.

[15]  Yang Xie,et al.  A community computational challenge to predict the activity of pairs of compounds , 2014, Nature Biotechnology.

[16]  Geoffrey E. Hinton,et al.  ImageNet classification with deep convolutional neural networks , 2012, Commun. ACM.

[17]  Sepp Hochreiter,et al.  Self-Normalizing Neural Networks , 2017, NIPS.

[18]  Yair Benita,et al.  An Unbiased Oncology Compound Screen to Identify Novel Combination Strategies , 2016, Molecular Cancer Therapeutics.

[19]  C. I. Bliss THE TOXICITY OF POISONS APPLIED JOINTLY1 , 1939 .

[20]  Krister Wennerberg,et al.  Methods for High-Throughput Drug Combination Screening and Synergy Scoring , 2016, bioRxiv.

[21]  Y. Kano,et al.  In vitro schedule-dependent interaction between paclitaxel and SN-38 (the active metabolite of irinotecan) in human carcinoma cell lines , 1998, Cancer Chemotherapy and Pharmacology.

[22]  K. Adachi,et al.  Schedule-dependent interaction between paclitaxel and 5-fluorouracil in human carcinoma cell lines in vitro. , 1996, British Journal of Cancer.

[23]  Pranita D. Tamma,et al.  Combination Therapy for Treatment of Infections with Gram-Negative Bacteria , 2012, Clinical Microbiology Reviews.

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

[25]  Xin Chen,et al.  DCDB: Drug combination database , 2010, Bioinform..

[26]  Dong-Sheng Cao,et al.  ChemoPy: freely available python package for computational biology and chemoinformatics , 2013, Bioinform..

[27]  S. Guichard,et al.  Sequence‐dependent activity of the irinotecan‐5FU combination in human colon‐cancer model HT‐29 in vitro and in vivo , 1997 .

[28]  Corinna Cortes,et al.  Support-Vector Networks , 1995, Machine Learning.

[29]  S. Loewe The problem of synergism and antagonism of combined drugs. , 1953, Arzneimittel-Forschung.

[30]  Mike Tyers,et al.  Prediction of Synergism from Chemical-Genetic Interactions by Machine Learning. , 2015, Cell systems.

[31]  L Li,et al.  A New Drug Combinatory Effect Prediction Algorithm on the Cancer Cell Based on Gene Expression and Dose–Response Curve , 2015, CPT: pharmacometrics & systems pharmacology.

[32]  E. De Clercq,et al.  The design of drugs for HIV and HCV. , 2007, Nature reviews. Drug discovery.

[33]  S. Guichard,et al.  Combination of oxaliplatin and irinotecan on human colon cancer cell lines: activity in vitro and in vivo , 2001, Anti-cancer drugs.

[34]  Andreas Bender,et al.  Modelling of compound combination effects and applications to efficacy and toxicity: state-of-the-art, challenges and perspectives. , 2016, Drug discovery today.

[35]  Raquel Chaves,et al.  The Importance of Cancer Cell Lines as in vitro Models in Cancer Methylome Analysis and Anticancer Drugs Testing , 2013 .

[36]  Tara N. Sainath,et al.  Deep convolutional neural networks for LVCSR , 2013, 2013 IEEE International Conference on Acoustics, Speech and Signal Processing.

[37]  Seta Shahin,et al.  A randomized phase IIIB trial of chemotherapy, bevacizumab, and panitumumab compared with chemotherapy and bevacizumab alone for metastatic colorectal cancer. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[39]  Ruth Nussinov,et al.  Structure and dynamics of molecular networks: A novel paradigm of drug discovery. A comprehensive review , 2012, Pharmacology & therapeutics.

[40]  Weimin Fan,et al.  Fulvestrant reverses doxorubicin resistance in multidrug-resistant breast cell lines independent of estrogen receptor expression , 2016, Oncology reports.

[41]  Jorge Cortes,et al.  Systems approaches and algorithms for discovery of combinatorial therapies. , 2009, Wiley interdisciplinary reviews. Systems biology and medicine.

[42]  Y Kano,et al.  Schedule-dependent synergism and antagonism between paclitaxel and methotrexate in human carcinoma cell lines. , 1998, Oncology research.

[43]  S. Hochreiter,et al.  DeepTox: Toxicity prediction using deep learning , 2017 .

[44]  Linda Mol,et al.  Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer. , 2009, The New England journal of medicine.

[45]  Y. Kano,et al.  Schedule-dependent interactions between paclitaxel and etoposide in human carcinoma cell lines in vitro , 1999, Cancer Chemotherapy and Pharmacology.

[46]  Xing Chen,et al.  NLLSS: Predicting Synergistic Drug Combinations Based on Semi-supervised Learning , 2016, PLoS Comput. Biol..

[47]  Jun Cui,et al.  Modeling of signaling crosstalk-mediated drug resistance and its implications on drug combination , 2016, Oncotarget.

[48]  David Rogers,et al.  Extended-Connectivity Fingerprints , 2010, J. Chem. Inf. Model..

[49]  STC Wong,et al.  DIGRE: Drug-Induced Genomic Residual Effect Model for Successful Prediction of Multidrug Effects , 2015, CPT: pharmacometrics & systems pharmacology.

[50]  Gaël Varoquaux,et al.  Scikit-learn: Machine Learning in Python , 2011, J. Mach. Learn. Res..

[51]  Klaus Obermayer,et al.  A new summarization method for affymetrix probe level data , 2006, Bioinform..

[52]  Jianxin Chen,et al.  Large-scale exploration and analysis of drug combinations , 2015, Bioinform..

[53]  L. Siu,et al.  Approaches to modernize the combination drug development paradigm , 2016, Genome Medicine.

[54]  Tara N. Sainath,et al.  FUNDAMENTAL TECHNOLOGIES IN MODERN SPEECH RECOGNITION Digital Object Identifier 10.1109/MSP.2012.2205597 , 2012 .

[55]  Anne E Carpenter,et al.  Repurposing High-Throughput Image Assays Enables Biological Activity Prediction for Drug Discovery. , 2018, Cell chemical biology.

[56]  J. Schellens,et al.  Increased oral bioavailability of topotecan in combination with the breast cancer resistance protein and P-glycoprotein inhibitor GF120918. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[57]  Xiaohua Ma,et al.  Mechanisms of drug combinations: interaction and network perspectives , 2009, Nature Reviews Drug Discovery.

[58]  Dennis Wang,et al.  Combenefit: an interactive platform for the analysis and visualization of drug combinations , 2016, Bioinform..

[59]  Camille Couprie,et al.  Learning Hierarchical Features for Scene Labeling , 2013, IEEE Transactions on Pattern Analysis and Machine Intelligence.

[60]  Y. Kano,et al.  Schedule‐dependent interactions between vinorelbine and paclitaxel in human carcinoma cell lines in vitro , 1999, Breast Cancer Research and Treatment.

[61]  Chris Morley,et al.  Open Babel: An open chemical toolbox , 2011, J. Cheminformatics.

[62]  Hans-Joachim Böhm,et al.  A guide to drug discovery: Hit and lead generation: beyond high-throughput screening , 2003, Nature Reviews Drug Discovery.

[63]  Geoffrey E. Hinton,et al.  Rectified Linear Units Improve Restricted Boltzmann Machines , 2010, ICML.

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

[65]  A. Groll,et al.  Recent advances in antifungal prevention and treatment. , 2009, Seminars in hematology.

[66]  MK Morris,et al.  Systematic Analysis of Quantitative Logic Model Ensembles Predicts Drug Combination Effects on Cell Signaling Networks , 2016, CPT: pharmacometrics & systems pharmacology.

[67]  R. E. White,et al.  High-throughput screening in drug metabolism and pharmacokinetic support of drug discovery. , 2000, Annual review of pharmacology and toxicology.

[68]  Leo Breiman,et al.  Random Forests , 2001, Machine Learning.

[69]  Erik De Clercq,et al.  The design of drugs for HIV and HCV , 2007, Nature Reviews Drug Discovery.

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

[71]  Hinrich W. H. Göhlmann,et al.  I/NI-calls for the exclusion of non-informative genes: a highly effective filtering tool for microarray data , 2007, Bioinform..

[72]  Pawan Kumar Gupta,et al.  Toxicophore exploration as a screening technology for drug design and discovery: techniques, scope and limitations , 2015, Archives of Toxicology.

[73]  Xing Chen,et al.  ASDCD: Antifungal Synergistic Drug Combination Database , 2014, PloS one.

[74]  J. Friedman Greedy function approximation: A gradient boosting machine. , 2001 .

[75]  Andreas Zell,et al.  jCompoundMapper: An open source Java library and command-line tool for chemical fingerprints , 2011, J. Cheminformatics.

[76]  Tapio Pahikkala,et al.  Toward more realistic drug^target interaction predictions , 2014 .