A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity.

Genes encoding components of the PI3K-AKT-mTOR signaling axis are frequently mutated in cancer, but few mutations have been characterized in MTOR, the gene encoding the mTOR kinase. Using publicly available tumor genome sequencing data, we generated a comprehensive catalog of mTOR pathway mutations in cancer, identifying 33 MTOR mutations that confer pathway hyperactivation. The mutations cluster in six distinct regions in the C-terminal half of mTOR and occur in multiple cancer types, with one cluster particularly prominent in kidney cancer. The activating mutations do not affect mTOR complex assembly, but a subset reduces binding to the mTOR inhibitor DEPTOR. mTOR complex 1 (mTORC1) signaling in cells expressing various activating mutations remains sensitive to pharmacologic mTOR inhibition, but is partially resistant to nutrient deprivation. Finally, cancer cell lines with hyperactivating MTOR mutations display heightened sensitivity to rapamycin both in culture and in vivo xenografts, suggesting that such mutations confer mTOR pathway dependency.

[1]  M. Tomita,et al.  Quantitative metabolome profiling of colon and stomach cancer microenvironment by capillary electrophoresis time-of-flight mass spectrometry. , 2009, Cancer research.

[2]  Laurie D. Smith,et al.  Exome Sequencing Reveals De Novo Germline Mutation of the Mammalian Target of Rapamycin (MTOR) in a Patient with Megalencephaly and Intractable Seizures , 2013 .

[3]  N. Pavletich,et al.  mTOR kinase structure, mechanism and regulation by the rapamycin-binding domain , 2013, Nature.

[4]  R. Arceci Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing , 2012 .

[5]  Suzanne Schubbert,et al.  Hyperactive Ras in developmental disorders and cancer , 2007, Nature Reviews Cancer.

[6]  D. Sabatini,et al.  DEPTOR Is an mTOR Inhibitor Frequently Overexpressed in Multiple Myeloma Cells and Required for Their Survival , 2009, Cell.

[7]  H. Aburatani,et al.  Genotype-dependent efficacy of a dual PI3K/mTOR inhibitor, NVP-BEZ235, and an mTOR inhibitor, RAD001, in endometrial carcinomas. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[8]  A. Lustig,et al.  Coupling of Glucose Deprivation with Impaired Histone H2B Monoubiquitination in Tumors , 2012, PloS one.

[9]  S. Shirasawa,et al.  Sorafenib enhances the therapeutic efficacy of rapamycin in colorectal cancers harboring oncogenic KRAS and PIK3CA. , 2012, Carcinogenesis.

[10]  S. Gabriel,et al.  Activating mTOR mutations in a patient with an extraordinary response on a phase I trial of everolimus and pazopanib. , 2014, Cancer discovery.

[11]  M. Loda,et al.  The oncogenic properties of mutant p110α and p110β phosphatidylinositol 3-kinases in human mammary epithelial cells , 2005 .

[12]  S. Gabriel,et al.  Discovery and saturation analysis of cancer genes across 21 tumor types , 2014, Nature.

[13]  T. Choueiri,et al.  Precision medicine for metastatic renal cell carcinoma. , 2014, Urologic oncology.

[14]  J. Urano,et al.  Point mutations in TOR confer Rheb-independent growth in fission yeast and nutrient-independent mammalian TOR signaling in mammalian cells , 2007, Proceedings of the National Academy of Sciences.

[15]  Steven J. M. Jones,et al.  Complete genomic landscape of a recurring sporadic parathyroid carcinoma , 2013, The Journal of pathology.

[16]  D. Sabatini,et al.  Amino acids and mTORC 1 : from lysosomes to disease , 2012 .

[17]  David Dunson,et al.  Genetic heterogeneity of diffuse large B-cell lymphoma , 2013, Proceedings of the National Academy of Sciences.

[18]  G. Mills,et al.  PIK3CA/PTEN Mutations and Akt Activation As Markers of Sensitivity to Allosteric mTOR Inhibitors , 2012, Clinical Cancer Research.

[19]  Gordon B Mills,et al.  mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. , 2006, Cancer research.

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

[21]  Steven J. M. Jones,et al.  Comprehensive molecular characterization of clear cell renal cell carcinoma , 2013, Nature.

[22]  Roberto Zoncu,et al.  Amino acids and mTORC1: from lysosomes to disease. , 2012, Trends in molecular medicine.

[23]  S. Gabriel,et al.  De novo somatic mutations in components of the PI3K-AKT3-mTOR pathway cause hemimegalencephaly , 2012, Nature Genetics.

[24]  A. Nakashima,et al.  Single amino-acid changes that confer constitutive activation of mTOR are discovered in human cancer , 2010, Oncogene.

[25]  S. Snyder,et al.  RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E-BP1. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[26]  D. Sabatini,et al.  mTOR Signaling in Growth Control and Disease , 2012, Cell.

[27]  Gary D Bader,et al.  International network of cancer genome projects , 2010, Nature.

[28]  D. Sabatini,et al.  mTOR: from growth signal integration to cancer, diabetes and ageing , 2010, Nature Reviews Molecular Cell Biology.

[29]  A. Marks,et al.  Rapamycin resistance is linked to defective regulation of Skp2. , 2012, Cancer research.

[30]  J. Blenis,et al.  Dominant mutations confer resistance to the immunosuppressant, rapamycin, in variants of a T cell lymphoma. , 1995, Cellular immunology.

[31]  H. Aburatani,et al.  Integrated molecular analysis of clear-cell renal cell carcinoma , 2013, Nature Genetics.

[32]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.

[33]  M. Birtwistle,et al.  Novel Somatic Mutations to PI3K Pathway Genes in Metastatic Melanoma , 2012, PloS one.

[34]  Benjamin E. Gross,et al.  The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. , 2012, Cancer discovery.

[35]  Elias Campo Guerri,et al.  International network of cancer genome projects , 2010 .

[36]  D. Sabatini,et al.  Discovery of 1-(4-(4-propionylpiperazin-1-yl)-3-(trifluoromethyl)phenyl)-9-(quinolin-3-yl)benzo[h][1,6]naphthyridin-2(1H)-one as a highly potent, selective mammalian target of rapamycin (mTOR) inhibitor for the treatment of cancer. , 2010, Journal of medicinal chemistry.

[37]  Li Zhao,et al.  Phosphatidylinositol 3-kinase: the oncoprotein. , 2010, Current topics in microbiology and immunology.

[38]  Matthew Meyerson,et al.  A Tumor Suppressor Complex with GAP Activity for the Rag GTPases That Signal Amino Acid Sufficiency to mTORC1 , 2013, Science.

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

[40]  M. Loda,et al.  The oncogenic properties of mutant p110alpha and p110beta phosphatidylinositol 3-kinases in human mammary epithelial cells. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[41]  David M. Sabatini,et al.  The Rag GTPases Bind Raptor and Mediate Amino Acid Signaling to mTORC1 , 2008, Science.

[42]  M. Hall,et al.  Rapamycin passes the torch: a new generation of mTOR inhibitors , 2011, Nature Reviews Drug Discovery.

[43]  Steven J. M. Jones,et al.  Comprehensive molecular characterization of clear cell renal cell carcinoma , 2013, Nature.

[44]  Mingming Jia,et al.  COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer , 2010, Nucleic Acids Res..

[45]  S. Snyder,et al.  RAFT 1 phosphorylation of the translational regulators p 70 S 6 kinase and 4 EBP 1 , 1998 .

[46]  Robert T. Jones,et al.  Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations , 2013, Nature Genetics.