This project seeks to exploit established mouse PDAC models/alleles, a strong oncogenomics and validation infrastructure, and a comprehensive PDAC genomics database in order to elucidate how signature genetic mutations contribute to the initiation and progression of PDAC and to discover and validate new PDAC oncogenes residing in regions of recurrent focal amplications. This project is driven by the hypothesis that the development of effective targeted therapies demands a comprehensive understanding of how known and novel lesions interact and influence tumor phenotypes. In the context of the established K-Ras-driven PDAC model, Project 1 will examine systematically the role of Ink4a/Arf, p53 and/or Smad4 mutations. All of the projects will work as a cohesive unit to determine the impact of each of these mutations on tumor histopathologic progression, invasive and metastatic potential (this project), angiogenic and stromal response (project 3), signaling pathway activation (project 2), and status of the cancer stem cell compartment (4). The efforts of Project 1 to reconstruct the key genetic events in the mouse will not only, build a PDAC progression model in which genotype-phenotype correlations can be established, but will also establish a modeling foundation upon which roles of yet-to-be discovered PDAC oncogenes can be validated in vivo. Indeed, it has become evident that many PDAC-relevant genes are emerging from ongoing genome scanning and transcriptome profiling efforts. Project 1 will make use of a large high-resolution human PDAC genomic database (including genomic profiles of our mouse tumors) and a battery of established functional and biochemical assays to begin to identify and validate novel PDAC oncogenes. As an illustrative example, Project 1 will pursue a systematic discovery and validation approach to the genes residing within a highly recurrent and focal amplicon with potential relevance to the PI3K pathway. The assays utilized emphasize the target's prevalence on human PDAC samples (in collaboration with the Experimental Pathology and Biobank Cores), its linkage to critical cancer cell signaling pathways (particularly Ras-PI3K in collaboration with Project 2), its expression/activity state in the cancer stem cell compartment (in collaboration with Project 4), and its role in PDAC tumor biology (particularly in shaping/maintaining the tumor microenvironment in collaboration with Project 3). Finally, we develop an inducible system for the P01 in which genotypephenotype of known and yet-to-be-discovered PDAC oncogenes can be elucidated.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA117969-05
Application #
8015366
Study Section
Subcommittee G - Education (NCI)
Project Start
Project End
Budget Start
2010-01-01
Budget End
2010-12-31
Support Year
5
Fiscal Year
2010
Total Cost
$221,884
Indirect Cost
Name
Dana-Farber Cancer Institute
Department
Type
DUNS #
076580745
City
Boston
State
MA
Country
United States
Zip Code
02215
Lundquist, Mark R; Goncalves, Marcus D; Loughran, Ryan M et al. (2018) Phosphatidylinositol-5-Phosphate 4-Kinases Regulate Cellular Lipid Metabolism By Facilitating Autophagy. Mol Cell 70:531-544.e9
Hopkins, Benjamin D; Pauli, Chantal; Du, Xing et al. (2018) Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature 560:499-503
Biancur, Douglas E; Kimmelman, Alec C (2018) The plasticity of pancreatic cancer metabolism in tumor progression and therapeutic resistance. Biochim Biophys Acta Rev Cancer 1870:67-75
Chen, Yang; LeBleu, Valerie S; Carstens, Julienne L et al. (2018) Dual reporter genetic mouse models of pancreatic cancer identify an epithelial-to-mesenchymal transition-independent metastasis program. EMBO Mol Med 10:
Hill, Margaret A; Alexander, William B; Guo, Bing et al. (2018) Kras and Tp53 Mutations Cause Cholangiocyte- and Hepatocyte-Derived Cholangiocarcinoma. Cancer Res 78:4445-4451
Mendt, Mayela; Kamerkar, Sushrut; Sugimoto, Hikaru et al. (2018) Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight 3:
Patra, Krushna C; Kato, Yasutaka; Mizukami, Yusuke et al. (2018) Mutant GNAS drives pancreatic tumourigenesis by inducing PKA-mediated SIK suppression and reprogramming lipid metabolism. Nat Cell Biol 20:811-822
Anglin, Justin; Zavareh, Reza Beheshti; Sander, Philipp N et al. (2018) Discovery and optimization of aspartate aminotransferase 1 inhibitors to target redox balance in pancreatic ductal adenocarcinoma. Bioorg Med Chem Lett 28:2675-2678
Yang, Annan; Herter-Sprie, Grit; Zhang, Haikuo et al. (2018) Autophagy Sustains Pancreatic Cancer Growth through Both Cell-Autonomous and Nonautonomous Mechanisms. Cancer Discov 8:276-287
Santana-Codina, Naiara; Roeth, Anjali A; Zhang, Yi et al. (2018) Oncogenic KRAS supports pancreatic cancer through regulation of nucleotide synthesis. Nat Commun 9:4945

Showing the most recent 10 out of 134 publications