Targeting mTOR dependency in pancreatic cancer

Objective Pancreatic cancer is a leading cause of cancer-related death in the Western world. Current chemotherapy regimens have modest survival benefit. Thus, novel, effective therapies are required for treatment of this disease. Design Activating KRAS mutation almost always drives pancreatic tumour initiation, however, deregulation of other potentially druggable pathways promotes tumour progression. PTEN loss leads to acceleration of KrasG12D-driven pancreatic ductal adenocarcinoma (PDAC) in mice and these tumours have high levels of mammalian target of rapamycin (mTOR) signalling. To test whether these KRAS PTEN pancreatic tumours show mTOR dependence, we compared response to mTOR inhibition in this model, to the response in another established model of pancreatic cancer, KRAS P53. We also assessed whether there was a subset of pancreatic cancer patients who may respond to mTOR inhibition. Results We found that tumours in KRAS PTEN mice exhibit a remarkable dependence on mTOR signalling. In these tumours, mTOR inhibition leads to proliferative arrest and even tumour regression. Further, we could measure response using clinically applicable positron emission tomography imaging. Importantly, pancreatic tumours driven by activated KRAS and mutant p53 did not respond to treatment. In human tumours, approximately 20% of cases demonstrated low PTEN expression and a gene expression signature that overlaps with murine KRAS PTEN tumours. Conclusions KRAS PTEN tumours are uniquely responsive to mTOR inhibition. Targeted anti-mTOR therapies may offer clinical benefit in subsets of human PDAC selected based on genotype, that are dependent on mTOR signalling. Thus, the genetic signatures of human tumours could be used to direct pancreatic cancer treatment in the future.

[1]  R. Rad,et al.  Selective requirement of PI3K/PDK1 signaling for Kras oncogene-driven pancreatic cell plasticity and cancer. , 2013, Cancer cell.

[2]  J. Asara,et al.  Stimulation of de Novo Pyrimidine Synthesis by Growth Signaling Through mTOR and S6K1 , 2013, Science.

[3]  U. Sauer,et al.  Quantitative Phosphoproteomics Reveal mTORC1 Activates de Novo Pyrimidine Synthesis , 2013, Science.

[4]  A. Maitra,et al.  Phosphorylation of ribosomal protein S6 attenuates DNA damage and tumor suppression during development of pancreatic cancer. , 2013, Cancer research.

[5]  N. Sonenberg,et al.  eIF4E/4E-BP ratio predicts the efficacy of mTOR targeted therapies. , 2012, Cancer Research.

[6]  Lincoln D. Stein,et al.  Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes , 2012, Nature.

[7]  G. Kristiansen,et al.  The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma , 2012, Nature.

[8]  Carlos Cuevas,et al.  Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. , 2012, Cancer cell.

[9]  A. Rust,et al.  Sleeping Beauty mutagenesis reveals cooperating mutations and pathways in pancreatic adenocarcinoma , 2012, Proceedings of the National Academy of Sciences.

[10]  D. Tuveson,et al.  nab-Paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse model of pancreatic cancer. , 2012, Cancer discovery.

[11]  J. Norman,et al.  Rab25 and CLIC3 Collaborate to Promote Integrin Recycling from Late Endosomes/Lysosomes and Drive Cancer Progression , 2012, Developmental cell.

[12]  David Olmos,et al.  First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[13]  Jennifer P Morton,et al.  MicroRNA Molecular Profiles Associated with Diagnosis, Clinicopathologic Criteria, and Overall Survival in Patients with Resectable Pancreatic Ductal Adenocarcinoma , 2011, Clinical Cancer Research.

[14]  Gerald C. Chu,et al.  PTEN is a major tumor suppressor in pancreatic ductal adenocarcinoma and regulates an NF-κB-cytokine network. , 2011, Cancer discovery.

[15]  Pierre Michel,et al.  FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. , 2011, The New England journal of medicine.

[16]  Rugang Zhang,et al.  Activation of the PIK3CA/AKT pathway suppresses senescence induced by an activated RAS oncogene to promote tumorigenesis. , 2011, Molecular cell.

[17]  D. Richel,et al.  mTOR inhibitor treatment of pancreatic cancer in a patient With Peutz-Jeghers syndrome. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[18]  M. Nowak,et al.  Distant Metastasis Occurs Late during the Genetic Evolution of Pancreatic Cancer , 2010, Nature.

[19]  A. Ashworth,et al.  LKB1 Haploinsufficiency Cooperates With Kras to Promote Pancreatic Cancer Through Suppression of p21-Dependent Growth Arrest , 2010, Gastroenterology.

[20]  R. Wolff,et al.  Inhibition of the mammalian target of rapamycin (mTOR) in advanced pancreatic cancer: results of two phase II studies , 2010, BMC Cancer.

[21]  N. Sonenberg,et al.  mTORC1-Mediated Cell Proliferation, But Not Cell Growth, Controlled by the 4E-BPs , 2010, Science.

[22]  Paul Timpson,et al.  Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer , 2010, Proceedings of the National Academy of Sciences.

[23]  A. Kibel Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial , 2009 .

[24]  N. Sonenberg,et al.  p53-dependent translational control of senescence and transformation via 4E-BPs. , 2009, Cancer cell.

[25]  David Allard,et al.  Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer , 2009, Science.

[26]  D. Guertin,et al.  The Pharmacology of mTOR Inhibition , 2009, Science Signaling.

[27]  Jeffrey W. Clark,et al.  Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[29]  Wei Chen,et al.  Predicting treatment response of malignant gliomas to bevacizumab and irinotecan by imaging proliferation with [18F] fluorothymidine positron emission tomography: a pilot study. , 2007, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[30]  Wolfgang A Weber,et al.  PET to assess early metabolic response and to guide treatment of adenocarcinoma of the oesophagogastric junction: the MUNICON phase II trial. , 2007, The Lancet. Oncology.

[31]  A. Toker Faculty Opinions recommendation of mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. , 2006 .

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

[33]  Huajun Yan,et al.  Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade , 2005, Molecular Cancer Therapeutics.

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

[35]  R. Hruban,et al.  Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. , 2005, Cancer cell.

[36]  E. Petricoin,et al.  Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. , 2003, Cancer cell.

[37]  S. Gambhir Molecular imaging of cancer with positron emission tomography , 2002, Nature Reviews Cancer.

[38]  G. Koehl,et al.  Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor , 2002, Nature Medicine.

[39]  R H Hruban,et al.  Progression model for pancreatic cancer. , 2000, Clinical cancer research : an official journal of the American Association for Cancer Research.

[40]  Otto Muzik,et al.  Imaging proliferation in vivo with [F-18]FLT and positron emission tomography , 1998, Nature Medicine.

[41]  D. V. Von Hoff,et al.  Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. , 1997, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[42]  A. Gingras,et al.  Rapamycin blocks the phosphorylation of 4E‐BP1 and inhibits cap‐dependent initiation of translation. , 1996, The EMBO journal.

[43]  N. Sigal,et al.  The immunosuppressive macrolides FK-506 and rapamycin act as reciprocal antagonists in murine T cells. , 1990, Journal of immunology.

[44]  D. Shibata,et al.  Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes , 1988, Cell.

[45]  G. Kristiansen,et al.  The deubiquitinase USP 9 X suppresses pancreatic ductal adenocarcinoma , 2012 .

[46]  A. Stütz Immunosuppressive macrolides. , 1992, Transplantation proceedings.