Bioinformatics Approaches to Predict Drug Responses from Genomic Sequencing.

Fulfilling the promises of precision medicine will depend on our ability to create patient-specific treatment regimens. Therefore, being able to translate genomic sequencing into predicting how a patient will respond to a given drug is critical. In this chapter, we review common bioinformatics approaches that aim to use sequencing data to predict sample-specific drug susceptibility. First, we explain the importance of customized drug regimens to the future of medical care. Second, we discuss the different public databases and community efforts that can be leveraged to develop new methods for identifying new predictive biomarkers. Third, we cover the basic methods that are currently used to identify markers or signatures of drug response, without any prior knowledge of the drug's mechanism of action. We further discuss how one can integrate knowledge about drug targets, mechanisms, and predictive markers to better estimate drug response in a diverse set of samples. We begin this section with a primer on popular methods to identify targets and mechanism of action for new small molecules. This discussion also includes a set of computational methods that incorporate other drug features, which do not relate to drug-induced genetic changes or sequencing data such as drug structures, side-effects, and efficacy profiles. Those additional drug properties can aid in gaining higher accuracy for the identification of drug target and mechanism of action. We then progress to discuss using these targets in combination with disease-specific expression patterns, known pathways, and genetic interaction networks to aid drug choice. Finally, we conclude this chapter with a general overview of machine learning methods that can integrate multiple pieces of sequencing data along with prior drug or biological knowledge to drastically improve response prediction.

[1]  J. Larkin,et al.  BRIM-1, -2 and -3 trials: improved survival with vemurafenib in metastatic melanoma patients with a BRAF(V600E) mutation. , 2012, Future oncology.

[2]  D. Botstein,et al.  A gene expression database for the molecular pharmacology of cancer , 2000, Nature Genetics.

[3]  S. Chanock,et al.  Analysis of chemotherapeutic response in ovarian cancers using publicly available high-throughput data. , 2014, Cancer research.

[4]  W. Huber,et al.  which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. MAnorm: a robust model for quantitative comparison of ChIP-Seq data sets , 2011 .

[5]  M. McCarthy,et al.  Genome-wide association studies for complex traits: consensus, uncertainty and challenges , 2008, Nature Reviews Genetics.

[6]  Manuel Hidalgo,et al.  Coordinated epidermal growth factor receptor pathway gene overexpression predicts epidermal growth factor receptor inhibitor sensitivity in pancreatic cancer. , 2008, Cancer research.

[7]  Paul A Clemons,et al.  The Connectivity Map: Using Gene-Expression Signatures to Connect Small Molecules, Genes, and Disease , 2006, Science.

[8]  Thomas Lengauer,et al.  A fast flexible docking method using an incremental construction algorithm. , 1996, Journal of molecular biology.

[9]  Helga Thorvaldsdóttir,et al.  Molecular signatures database (MSigDB) 3.0 , 2011, Bioinform..

[10]  J. Briones Emerging therapies for B-cell non-Hodgkin lymphoma , 2009, Expert review of anticancer therapy.

[11]  R. Shoemaker The NCI60 human tumour cell line anticancer drug screen , 2006, Nature Reviews Cancer.

[12]  Eytan Ruppin,et al.  Predicting Cancer-Specific Vulnerability via Data-Driven Detection of Synthetic Lethality , 2014, Cell.

[13]  A. Giaccia,et al.  Harnessing synthetic lethal interactions in anticancer drug discovery , 2011, Nature Reviews Drug Discovery.

[14]  L. Staudt,et al.  Small molecule inhibitors of IκB kinase are selectively toxic for subgroups of diffuse large B-cell lymphoma defined by gene expression profiling , 2005 .

[15]  Chartchalerm Isarankura-Na-Ayudhya,et al.  Advances in computational methods to predict the biological activity of compounds , 2010, Expert opinion on drug discovery.

[16]  C. Ting,et al.  A genome-wide association study identifies new loci for ACE activity: potential implications for response to ACE inhibitor , 2010, The Pharmacogenomics Journal.

[17]  S. Ramaswamy,et al.  Systematic identification of genomic markers of drug sensitivity in cancer cells , 2012, Nature.

[18]  L. Samson,et al.  Genomic predictors of interindividual differences in response to DNA damaging agents. , 2008, Genes & development.

[19]  Nan Song,et al.  Analysis of chemotherapeutic response heterogeneity and drug clustering based on mechanism of action using an in vitro assay. , 2010, Anticancer research.

[20]  Neel Madhukar,et al.  Abstract B162: Small molecule target prediction and identification of novel anti-cancer compounds using a data-driven bayesian approach , 2015 .

[21]  Xiaomin Luo,et al.  TarFisDock: a web server for identifying drug targets with docking approach , 2006, Nucleic Acids Res..

[22]  Simon C. Potter,et al.  Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls , 2007, Nature.

[23]  Matthew R. Laird,et al.  Protein Protein Interaction Network Evaluation for Identifying Potential Drug Targets , 2009 .

[24]  S. Gabriel,et al.  EGFR Mutations in Lung Cancer: Correlation with Clinical Response to Gefitinib Therapy , 2004, Science.

[25]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[26]  Wentian Li,et al.  Genome-Wide Association Scan Identifies Candidate Polymorphisms Associated with Differential Response to Anti-TNF Treatment in Rheumatoid Arthritis , 2008, Molecular medicine.

[27]  F. Cappuzzo,et al.  HER2 mutation and response to trastuzumab therapy in non-small-cell lung cancer. , 2006, The New England journal of medicine.

[28]  P. Hirth,et al.  Vemurafenib: the first drug approved for BRAF-mutant cancer , 2012, Nature Reviews Drug Discovery.

[29]  Henning Hermjakob,et al.  The Reactome pathway Knowledgebase , 2015, Nucleic acids research.

[30]  David R. Kelley,et al.  Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks , 2012, Nature Protocols.

[31]  Mathias Wilhelm,et al.  Global proteome analysis of the NCI-60 cell line panel. , 2013, Cell reports.

[32]  Jason H. Moore,et al.  Chapter 11: Genome-Wide Association Studies , 2012, PLoS Comput. Biol..

[33]  Yudong D. He,et al.  Functional Discovery via a Compendium of Expression Profiles , 2000, Cell.

[34]  R. Collins,et al.  SLCO1B1 variants and statin-induced myopathy--a genomewide study. , 2008, The New England journal of medicine.

[35]  Nicholas M. Pajewski,et al.  Six Degrees of Epistasis: Statistical Network Models for GWAS , 2011, Front. Gene..

[36]  Karin Breuer,et al.  InnateDB: systems biology of innate immunity and beyond—recent updates and continuing curation , 2012, Nucleic Acids Res..

[37]  C. Orengo,et al.  Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma , 2006, BMC Genomics.

[38]  Lin He,et al.  Prediction of Drug-Target Interactions for Drug Repositioning Only Based on Genomic Expression Similarity , 2013, PLoS Comput. Biol..

[39]  E. Pearson,et al.  Insights from genome-wide association studies of drug response. , 2013, Annual review of pharmacology and toxicology.

[40]  D. Haussler,et al.  The Somatic Genomic Landscape of Glioblastoma , 2013, Cell.

[41]  Lyndsay N Harris,et al.  Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[42]  Susumu Goto,et al.  KEGG: Kyoto Encyclopedia of Genes and Genomes , 2000, Nucleic Acids Res..

[43]  J. Marchini,et al.  Genotype Imputation with Thousands of Genomes , 2011, G3: Genes | Genomes | Genetics.

[44]  D. Butina,et al.  Predicting ADME properties in silico: methods and models. , 2002, Drug discovery today.

[45]  Pablo Villoslada,et al.  Genome-wide pharmacogenomic analysis of the response to interferon beta therapy in multiple sclerosis. , 2008, Archives of neurology.

[46]  M. Ashburner,et al.  Gene Ontology: tool for the unification of biology , 2000, Nature Genetics.

[47]  M. Eileen Dolan,et al.  Chemotherapeutic drug susceptibility associated SNPs are enriched in expression quantitative trait loci , 2010, Proceedings of the National Academy of Sciences.

[48]  A. Hauschild,et al.  Improved survival with vemurafenib in melanoma with BRAF V600E mutation. , 2011, The New England journal of medicine.

[49]  Mary Regina Boland,et al.  A Computational Drug Repositioning Approach for Targeting Oncogenic Transcription Factors. , 2016, Cell reports.

[50]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[51]  Michiaki Kubo,et al.  Genome-Wide Association Study: A Useful Tool to Identify Common Genetic Variants Associated with Drug Toxicity and Efficacy in Cancer Pharmacogenomics , 2014, Clinical Cancer Research.

[52]  P. Bork,et al.  Drug Target Identification Using Side-Effect Similarity , 2008, Science.

[53]  Jack A. Taylor,et al.  SNPinfo: integrating GWAS and candidate gene information into functional SNP selection for genetic association studies , 2009, Nucleic Acids Res..

[54]  Julio Saez-Rodriguez,et al.  Machine Learning Prediction of Cancer Cell Sensitivity to Drugs Based on Genomic and Chemical Properties , 2012, PloS one.

[55]  Ellen T. Gelfand,et al.  The Genotype-Tissue Expression (GTEx) project , 2013, Nature Genetics.

[56]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[57]  P. Meltzer,et al.  NCI-60 Whole Exome Sequencing and Pharmacological CellMiner Analyses , 2014, PloS one.

[58]  Justin Lamb,et al.  The Connectivity Map: a new tool for biomedical research , 2007, Nature Reviews Cancer.

[59]  P. Meltzer,et al.  The exomes of the NCI-60 panel: a genomic resource for cancer biology and systems pharmacology. , 2013, Cancer research.

[60]  Olivier Elemento,et al.  A primer on precision medicine informatics , 2016, Briefings Bioinform..

[61]  Michael P. Morrissey,et al.  Pharmacogenomic agreement between two cancer cell line data sets , 2015, Nature.

[62]  Adrian Wiestner,et al.  A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma , 2003, Proceedings of the National Academy of Sciences of the United States of America.

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

[64]  Joel Dudley,et al.  Exploiting drug-disease relationships for computational drug repositioning , 2011, Briefings Bioinform..

[65]  A. Weeraratna,et al.  Gene expression response to cisplatin treatment in drug-sensitive and drug-resistant ovarian cancer cells , 2007, Oncogene.

[66]  B. Leber,et al.  Identification of Drugs Including a Dopamine Receptor Antagonist that Selectively Target Cancer Stem Cells , 2012, Cell.

[67]  Gaurav Pandey,et al.  Prediction of Genetic Interactions Using Machine Learning and Network Properties , 2015, Front. Bioeng. Biotechnol..

[68]  Greg Yothers,et al.  Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. , 2005, The New England journal of medicine.

[69]  Adam A. Margolin,et al.  The Cancer Cell Line Encyclopedia enables predictive modeling of anticancer drug sensitivity , 2012, Nature.

[70]  Manuel A. R. Ferreira,et al.  PLINK: a tool set for whole-genome association and population-based linkage analyses. , 2007, American journal of human genetics.