Discovering novel pharmacogenomic biomarkers by imputing drug response in cancer patients from large genomics studies.

Obtaining accurate drug response data in large cohorts of cancer patients is very challenging; thus, most cancer pharmacogenomics discovery is conducted in preclinical studies, typically using cell lines and mouse models. However, these platforms suffer from serious limitations, including small sample sizes. Here, we have developed a novel computational method that allows us to impute drug response in very large clinical cancer genomics data sets, such as The Cancer Genome Atlas (TCGA). The approach works by creating statistical models relating gene expression to drug response in large panels of cancer cell lines and applying these models to tumor gene expression data in the clinical data sets (e.g., TCGA). This yields an imputed drug response for every drug in each patient. These imputed drug response data are then associated with somatic genetic variants measured in the clinical cohort, such as copy number changes or mutations in protein coding genes. These analyses recapitulated drug associations for known clinically actionable somatic genetic alterations and identified new predictive biomarkers for existing drugs.

[1]  Francisco Azuaje,et al.  Computational models for predicting drug responses in cancer research , 2016, Briefings Bioinform..

[2]  Nancy J. Cox,et al.  Consistency in large pharmacogenomic studies , 2016, Nature.

[3]  Fupan Yao,et al.  Tissue specificity of in vitro drug sensitivity , 2016, bioRxiv.

[4]  N. Cox,et al.  Cancer biomarker discovery is improved by accounting for variability in general levels of drug sensitivity in pre-clinical models , 2016, Genome Biology.

[5]  N. Schultz,et al.  OncoKB: Annotation of the oncogenic effect and treatment implications of somatic mutations in cancer. , 2016 .

[6]  J. Mesirov,et al.  DiSCoVERing Innovative Therapies for Rare Tumors: Combining Genetically Accurate Disease Models with In Silico Analysis to Identify Novel Therapeutic Targets , 2016, Clinical Cancer Research.

[7]  Melissa C. Skala,et al.  Drug response in organoids generated from frozen primary tumor tissues , 2016, Scientific Reports.

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

[9]  M. Maitland,et al.  Predicting Response to Histone Deacetylase Inhibitors Using High-Throughput Genomics. , 2015, Journal of the National Cancer Institute.

[10]  Joshua M. Korn,et al.  High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response , 2015, Nature Medicine.

[11]  Large-Scale Drug Screens Support Precision Medicine. , 2015, Cancer discovery.

[12]  Joshua A. Bittker,et al.  Harnessing Connectivity in a Large-Scale Small-Molecule Sensitivity Dataset. , 2015, Cancer discovery.

[13]  William E. Evans,et al.  Pharmacogenomics in the clinic , 2015, Nature.

[14]  R. Finley,et al.  A novel ER–microtubule-binding protein, ERLIN2, stabilizes Cyclin B1 and regulates cell cycle progression , 2015, Cell Discovery.

[15]  Steffen Falgreen,et al.  Predicting response to multidrug regimens in cancer patients using cell line experiments and regularised regression models , 2015, BMC Cancer.

[16]  Qing Zhao,et al.  Combining multidimensional genomic measurements for predicting cancer prognosis: observations from TCGA , 2015, Briefings Bioinform..

[17]  R. Lanfear,et al.  The Extent and Consequences of P-Hacking in Science , 2015, PLoS biology.

[18]  Patrick Aloy,et al.  Drug sensitivity in cancer cell lines is not tissue-specific , 2015, Molecular Cancer.

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

[20]  Paul Geeleher,et al.  pRRophetic: An R Package for Prediction of Clinical Chemotherapeutic Response from Tumor Gene Expression Levels , 2014, PloS one.

[21]  S. Dudoit,et al.  Normalization of RNA-seq data using factor analysis of control genes or samples , 2014, Nature Biotechnology.

[22]  Benjamin J. Raphael,et al.  Multiplatform Analysis of 12 Cancer Types Reveals Molecular Classification within and across Tissues of Origin , 2014, Cell.

[23]  Adam A. Margolin,et al.  Assessing the clinical utility of cancer genomic and proteomic data across tumor types , 2014, Nature Biotechnology.

[24]  S. Barollo,et al.  The combination of RAF265, SB590885, ZSTK474 on thyroid cancer cell lines deeply impact on proliferation and MAPK and PI3K/Akt signaling pathways , 2014, Investigational New Drugs.

[25]  Robert L. Grossman,et al.  Bionimbus: a cloud for managing, analyzing and sharing large genomics datasets , 2014, J. Am. Medical Informatics Assoc..

[26]  D. Amadori,et al.  Palbociclib (PD 0332991): targeting the cell cycle machinery in breast cancer , 2014, Expert opinion on pharmacotherapy.

[27]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[28]  Laura M. Heiser,et al.  A community effort to assess and improve drug sensitivity prediction algorithms , 2014, Nature Biotechnology.

[29]  N. Cox,et al.  Clinical drug response can be predicted using baseline gene expression levels and in vitro drug sensitivity in cell lines , 2014, Genome Biology.

[30]  Benjamin Haibe-Kains,et al.  Inconsistency in large pharmacogenomic studies , 2013, Nature.

[31]  H. Hollema,et al.  Nutlin-3 preferentially sensitises wild-type p53-expressing cancer cells to DR5-selective TRAIL over rhTRAIL , 2013, British Journal of Cancer.

[32]  Gary D Bader,et al.  Comprehensive identification of mutational cancer driver genes across 12 tumor types , 2013, Scientific Reports.

[33]  S. Gabriel,et al.  Pan-cancer patterns of somatic copy-number alteration , 2013, Nature Genetics.

[34]  Joshua M. Stuart,et al.  The Cancer Genome Atlas Pan-Cancer analysis project , 2013, Nature Genetics.

[35]  Robert Gentleman,et al.  Software for Computing and Annotating Genomic Ranges , 2013, PLoS Comput. Biol..

[36]  X. Hua,et al.  Interaction of the oncogenic miR-21 microRNA and the p53 tumor suppressor pathway. , 2013, Carcinogenesis.

[37]  Benjamin E. Gross,et al.  Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal , 2013, Science Signaling.

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

[39]  N. Moatamed,et al.  High concordance between HercepTest immunohistochemistry and ERBB2 fluorescence in situ hybridization before and after implementation of American Society of Clinical Oncology/College of American Pathology 2007 guidelines , 2012, Modern Pathology.

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

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

[42]  Paul L Appleton,et al.  The microtubule poison vinorelbine kills cells independently of mitotic arrest and targets cells lacking the APC tumour suppressor more effectively , 2012, Journal of Cell Science.

[43]  G. Clayman,et al.  MEK Inhibitor PD0325901 Significantly Reduces the Growth of Papillary Thyroid Carcinoma Cells In vitro and In vivo , 2010, Molecular Cancer Therapeutics.

[44]  H. Allgayer,et al.  MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer , 2008, Oncogene.

[45]  J. Fletcher,et al.  Long-term results from a randomized phase II trial of standard- versus higher-dose imatinib mesylate for patients with unresectable or metastatic gastrointestinal stromal tumors expressing KIT. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[46]  K. Coombes,et al.  Microarrays: retracing steps , 2007, Nature Medicine.

[47]  Xin Huang,et al.  Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial , 2006, The Lancet.

[48]  M. Ostland,et al.  Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[49]  Patricia L. Harris,et al.  Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. , 2004, The New England journal of medicine.

[50]  J F Barrett,et al.  Identification of CDK4 as a target of c-MYC. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[51]  J. Bonneterre,et al.  Vinorelbine (navelbine) as a salvage treatment for advanced breast cancer. , 1994, Annals of oncology : official journal of the European Society for Medical Oncology.

[52]  D. Pinkel,et al.  ERBB2 amplification in breast cancer analyzed by fluorescence in situ hybridization. , 1992, Proceedings of the National Academy of Sciences of the United States of America.