Systematic analysis of genotype‐specific drug responses in cancer

A systematic understanding of genotype‐specific sensitivity or resistance to anticancer agents is required to provide improved patient therapy. The availability of an expansive panel of annotated cancer cell lines enables comparative surveys of associations between genotypes and compounds of various target classes. Thus, one can better predict the optimal treatment for a specific tumor. Here, we present a statistical framework, cell line enrichment analysis (CLEA), to associate the response of anticancer agents with major cancer genotypes. Multilevel omics data, including transcriptome, proteome and phosphatome data, were integrated with drug data based on the genotypic classification of cancer cell lines. The results reproduced known patterns of compound sensitivity associated with particular genotypes. In addition, this approach reveals multiple unexpected associations between compounds and mutational genotypes. The mutational genotypes led to unique protein activation and gene expression signatures, which provided a mechanistic understanding of their functional effects. Furthermore, CLEA maps revealed interconnections between TP53 mutations and other mutations in the context of drug responses. The TP53 mutational status appears to play a dominant role in determining clustering patterns of gene and protein expression profiles for major cancer genotypes. This study provides a framework for the integrative analysis of mutations, drug responses and omics data in cancers.

[1]  A. Iafrate,et al.  Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling , 2007, Proceedings of the National Academy of Sciences.

[2]  Joel Greshock,et al.  Molecular target class is predictive of in vitro response profile. , 2010, Cancer research.

[3]  H. McLeod,et al.  Pharmacogenomics: from bedside to clinical practice. , 2006, Human molecular genetics.

[4]  G. Prendergast,et al.  RhoB facilitates c-Myc turnover by supporting efficient nuclear accumulation of GSK-3 , 2006, Oncogene.

[5]  J. Settleman Cell culture modeling of genotype-directed sensitivity to selective kinase inhibitors: targeting the anaplastic lymphoma kinase (ALK). , 2009, Seminars in oncology.

[6]  G. Mills,et al.  Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells , 2006, Molecular Cancer Therapeutics.

[7]  William E. Evans,et al.  Acute lymphoblastic leukaemia: a model for the pharmacogenomics of cancer therapy , 2006, Nature Reviews Cancer.

[8]  H. Auer,et al.  Tissue-Wide Expression Profiling Using cDNA Subtraction and Microarrays to Identify Tumor-Specific Genes , 2004, Cancer Research.

[9]  D. Zecchin,et al.  Replacement of normal with mutant alleles in the genome of normal human cells unveils mutation-specific drug responses , 2008, Proceedings of the National Academy of Sciences.

[10]  Noor B. Dawany,et al.  Asymmetric microarray data produces gene lists highly predictive of research literature on multiple cancer types , 2010, BMC Bioinformatics.

[11]  F. D. De Braud,et al.  Pharmacogenetics of Anticancer Drug Sensitivity in Non-Small Cell Lung Cancer , 2003, Pharmacological Reviews.

[12]  Zhi Hu,et al.  An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. , 2008, Cancer research.

[13]  Pekka Tiikkainen,et al.  Comparison of structure fingerprint and molecular interaction field based methods in explaining biological similarity of small molecules in cell-based screens , 2009, J. Comput. Aided Mol. Des..

[14]  M. Hollstein,et al.  p53 and human cancer: the first ten thousand mutations. , 2000, Advances in cancer research.

[15]  P. Houghton,et al.  N-Myc induction stimulated by insulin-like growth factor I through mitogen-activated protein kinase signaling pathway in human neuroblastoma cells. , 2000, Cancer research.

[16]  D. Coulter,et al.  Inhibition of IGF-I receptor signaling in combination with rapamycin or temsirolimus increases MYC-N phosphorylation. , 2009, Anticancer research.

[17]  D. Carson,et al.  Cancer progression and p53 , 1995, The Lancet.

[18]  M. Stratton,et al.  The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website , 2004, British Journal of Cancer.

[19]  Yiling Lu,et al.  AKT-independent signaling downstream of oncogenic PIK3CA mutations in human cancer. , 2009, Cancer cell.

[20]  Michael Krauthammer,et al.  Structural similarity assessment for drug sensitivity prediction in cancer , 2009, BMC Bioinformatics.

[21]  A. Eklund,et al.  Prediction of drug efficacy for cancer treatment based on comparative analysis of chemosensitivity and gene expression data. , 2012, Bioorganic & medicinal chemistry.

[22]  Jae K. Lee,et al.  Transcript and protein expression profiles of the NCI-60 cancer cell panel: an integromic microarray study , 2007, Molecular Cancer Therapeutics.

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

[24]  A. Børresen-Dale,et al.  TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes , 2007, Oncogene.

[25]  Todd R. Golub,et al.  BRAF mutation predicts sensitivity to MEK inhibition , 2006, Nature.

[26]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.