Deregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimus.

Personalized cancer medicine is based on the concept that targeted therapies are effective on subsets of patients whose tumors carry specific molecular alterations. Several mammalian target of rapamycin (mTOR) inhibitors are in preclinical or clinical trials for cancers, but the molecular basis of sensitivity or resistance to these inhibitors among patients is largely unknown. Here we have identified oncogenic variants of phosphoinositide-3-kinase, catalytic, alpha polypeptide (PIK3CA) and KRAS as determinants of response to the mTOR inhibitor everolimus. Human cancer cells carrying alterations in the PI3K pathway were responsive to everolimus, both in vitro and in vivo, except when KRAS mutations occurred concomitantly or were exogenously introduced. In human cancer cells with mutations in both PIK3CA and KRAS, genetic ablation of mutant KRAS reinstated response to the drug. Consistent with these data, PIK3CA mutant cells, but not KRAS mutant cells, displayed everolimus-sensitive translation. Importantly, in a cohort of metastatic cancer patients, the presence of oncogenic KRAS mutations was associated with lack of benefit after everolimus therapy. Thus, our results demonstrate that alterations in the KRAS and PIK3CA genes may represent biomarkers to optimize treatment of patients with mTOR inhibitors.

[1]  J. Blenis,et al.  The RSK family of kinases: emerging roles in cellular signalling , 2008, Nature Reviews Molecular Cell Biology.

[2]  Funda Meric-Bernstam,et al.  Targeting the mTOR signaling network for cancer therapy. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[3]  Yiling Lu,et al.  Exploiting the PI3K/AKT Pathway for Cancer Drug Discovery , 2005, Nature Reviews Drug Discovery.

[4]  M. van Glabbeke,et al.  New guidelines to evaluate the response to treatment in solid tumors , 2000, Journal of the National Cancer Institute.

[5]  Umar Mahmood,et al.  Development of a mouse model for sporadic and metastatic colon tumors and its use in assessing drug treatment , 2010, Proceedings of the National Academy of Sciences.

[6]  A. Bardelli,et al.  Isogenic mutant human cells: A new tool for personalized cancer medicine , 2010, Cell cycle.

[7]  W. Ma,et al.  Novel Agents on the Horizon for Cancer Therapy , 2009, CA: a cancer journal for clinicians.

[8]  J. Blenis,et al.  RAS/ERK Signaling Promotes Site-specific Ribosomal Protein S6 Phosphorylation via RSK and Stimulates Cap-dependent Translation* , 2007, Journal of Biological Chemistry.

[9]  H. Lane,et al.  Identifying optimal biologic doses of everolimus (RAD001) in patients with cancer based on the modeling of preclinical and clinical pharmacokinetic and pharmacodynamic data. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[10]  Silvia Benvenuti,et al.  Gene copy number for epidermal growth factor receptor (EGFR) and clinical response to antiEGFR treatment in colorectal cancer: a cohort study. , 2005, The Lancet. Oncology.

[11]  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.

[12]  David McDermott,et al.  Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. , 2007, The New England journal of medicine.

[13]  R. Motzer,et al.  Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial , 2008, The Lancet.

[14]  Sridhar Ramaswamy,et al.  Synthetic Lethal Interaction between Oncogenic KRAS Dependency and STK33 Suppression in Human Cancer Cells , 2009, Cell.

[15]  M Van Glabbeke,et al.  New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. , 2000, Journal of the National Cancer Institute.

[16]  D. Xing,et al.  A genetically defined mouse ovarian carcinoma model for the molecular characterization of pathway-targeted therapy and tumor resistance. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[17]  L. Helman,et al.  Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism , 2007, Oncogene.

[18]  Philippe P Roux,et al.  Oncogenic MAPK Signaling Stimulates mTORC1 Activity by Promoting RSK-Mediated Raptor Phosphorylation , 2008, Current Biology.

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

[20]  F. Khuri,et al.  Enhancing mammalian target of rapamycin (mTOR)-targeted cancer therapy by preventing mTOR/raptor inhibition-initiated, mTOR/rictor-independent Akt activation. , 2008, Cancer research.

[21]  Suzanne F. Jones,et al.  Dose- and schedule-dependent inhibition of the mammalian target of rapamycin pathway with everolimus: a phase I tumor pharmacodynamic study in patients with advanced solid tumors. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[22]  A. Bardelli,et al.  Biomarkers Predicting Clinical Outcome of Epidermal Growth Factor Receptor–Targeted Therapy in Metastatic Colorectal Cancer , 2009, Journal of the National Cancer Institute.

[23]  S. Grosso,et al.  PKCbetaII modulates translation independently from mTOR and through RACK1. , 2008, The Biochemical journal.

[24]  S. Shirasawa,et al.  Altered growth of human colon cancer cell lines disrupted at activated Ki-ras. , 1993, Science.

[25]  J. Ptak,et al.  Colorectal cancer: Mutations in a signalling pathway , 2005, Nature.

[26]  F. Khuri,et al.  Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. , 2005, Cancer research.

[27]  J. Hershey,et al.  AGC kinases regulate phosphorylation and activation of eukaryotic translation initiation factor 4B , 2009, Oncogene.

[28]  M. Konopleva,et al.  Phase I/II Study of the Mammalian Target of Rapamycin Inhibitor Everolimus (RAD001) in Patients with Relapsed or Refractory Hematologic Malignancies , 2006, Clinical Cancer Research.

[29]  G. Toner Clinical relevance of "late" in the management of late relapse after treatment for a germ cell tumor. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[30]  Carlo Rago,et al.  Mutant PIK3CA promotes cell growth and invasion of human cancer cells. , 2005, Cancer cell.

[31]  R. McLendon,et al.  Phase 2 trial of erlotinib plus sirolimus in adults with recurrent glioblastoma , 2009, Journal of Neuro-Oncology.

[32]  R. Broaddus,et al.  A phase II study of oral mammalian target of rapamycin (mTOR) inhibitor, RAD001 (everolimus), in patients with recurrent endometrial carcinoma (EC) , 2008 .

[33]  Suzanne F. Jones,et al.  Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[35]  G. Thomas,et al.  mTOR Complex1-S6K1 signaling: at the crossroads of obesity, diabetes and cancer. , 2007, Trends in molecular medicine.

[36]  P. Pandolfi,et al.  Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. , 2008, The Journal of clinical investigation.

[37]  F. Cavalli,et al.  PTEN loss of expression predicts cetuximab efficacy in metastatic colorectal cancer patients , 2007, British Journal of Cancer.

[38]  J. Blenis,et al.  Molecular mechanisms of mTOR-mediated translational control , 2009, Nature Reviews Molecular Cell Biology.

[39]  G. Kroemer,et al.  Current development of mTOR inhibitors as anticancer agents , 2006, Nature Reviews Drug Discovery.