The therapeutic problem we address is how to inhibit the development and progression of IPMN (intraductal papillary mucinous neoplasm), PanIN (Pancreatic Intraepithelial Neoplasia) and PDA (pancreatic ductal adenocarcinoma). The biological problem is to identify key signaling pathways that initiate and propagate IPMN, PanIN and PDA. A central debate in the field is whether PDA initiates in pancreatic progenitor cells or with dedifferentiation of parenchymal acinar cells to progenitor-like ductal cells, in a process termed acinar to ductal metaplasia (ADM). The function of the transcription factor Master Regulators (MRs) of acinar cell identity is critical to this process but post-translational regulaton of their activity through receptor tyrosine kinase (RTK) and G protein coupled receptor (GPCR) signaling is completely unexplored. Regulators of G protein signaling (RGS) proteins integrate multiple intracellular signals from RTKs and GPCRs to feedback regulate Gi and Gq protein signaling. We have developed a GFP reporter (Rgs16:GFP) that allows us to monitor neoplasia and tumor formation from early stages in in vivo mouse models of IPMN, PanIN and PDA. Combining the Rgs16:GFP reporter with mouse models for IPMN, PanIN and PDA is providing insights into development of these tumors and a novel and rapid in vivo screen for inhibitors of this process. We discovered RGS genes that suppress progression of IPMN to invasive carcinoma. Based on preliminary analyses using these models, we find that IPMN and PanIN initiate in discrete ductal progenitor cells in response to stress in the acinar or endocrine compartments of the pancreas. This interpretation challenges the currently favored paradigm that PDA initiates with cells that underwent ADM whereas we propose that chronic ADM stimulates proliferation of pre-existing progenitor cells in ducts. We find that the RTK Axl is expressed in pancreatic ducts and is highly expressed with Rgs16:GFP in PDA progenitor cells in primary culture. Our hypothesis is that the Axl-ligand Gas6 expressed in stroma evokes Axl signaling in epithelial progenitor cells and contributes to activation of KrasG12D, post-translational control of MR levels and/or activity, PDA initiation and progression. In our preliminary in vivo screens of new therapeutics to inhibit PDA progression, we determined that warfarin, which blocks Gas6 induced activation of Axl, combined with the standard of care Gemcitabine and Abraxane (GA), significantly reduced PDA progression.
The Specific Aims of this proposal are 1) Identify optimal drug combinations, including inhibitors of Axl signaling, to inhibit PDA. 2) Identify early markers of IPMN, PanIN and PDA in plasma. 3) Determine Gas6-Axl function in PDA. The proposed project, if successful, will have a major impact on cancer by i) identifying early blood markers of PDA, and perhaps other epithelial cancers, ii) introducing a novel, general and rapid in vivo screening technique to find novel drug combinations that inhibit PDA progression, iii) providing novel genetic tools to determine if PDA initiates in progenitor cells in the pancreatic duct, Iv) identify previously unknown regulatory mechanisms of the MR transcription factors.

Public Health Relevance

to public health in this project is the exploitation of a novel rapid in vivo screen for effective therapies for pancreatic ductal adenocarcinoma (PDA). The screen is based on reporter gene expression that is dependent upon activation of Kras, the dominant oncogenic driver of PDA. We will determine the efficacy of rationally chosen therapies, document the mechanism of action of candidate therapies and use the in vivo model to identity potential markers of early disease.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA192381-05
Application #
9626896
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Alley, Michael C
Project Start
2015-02-10
Project End
2020-01-31
Budget Start
2019-02-01
Budget End
2020-01-31
Support Year
5
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of Texas Sw Medical Center Dallas
Department
Surgery
Type
Schools of Medicine
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
Du, Wenting; Brekken, Rolf A (2018) Does Axl have potential as a therapeutic target in pancreatic cancer? Expert Opin Ther Targets 22:955-966
Zhang, Yuqing; Kirane, Amanda; Huang, Huocong et al. (2018) Cyclooxygenase-2 inhibition potentiates the efficacy of vascular endothelial growth factor blockade and promotes an immune stimulatory microenvironment in preclinical models of pancreatic cancer. Mol Cancer Res :
Cruz, Victoria H; Arner, Emily N; Wynne, Katherine W et al. (2018) Loss of Tbk1 kinase activity protects mice from diet-induced metabolic dysfunction. Mol Metab 16:139-149
Zolghadri, Yalda; Pal Choudhuri, Shreoshi; Ocal, Ozhan et al. (2018) Malnutrition in Pancreatic Ductal Adenocarcinoma (PDA): Dietary Pancreatic Enzymes Improve Short-Term Health but Stimulate Tumor Growth. Am J Pathol 188:616-626
Ludwig, Kathleen F; Du, Wenting; Sorrelle, Noah B et al. (2018) Small-Molecule Inhibition of Axl Targets Tumor Immune Suppression and Enhances Chemotherapy in Pancreatic Cancer. Cancer Res 78:246-255
Du, Wenting; Huang, Huocong; Sorrelle, Noah et al. (2018) Sitravatinib potentiates immune checkpoint blockade in refractory cancer models. JCI Insight 3:
Wieczorek, Michal; Tcherkezian, Joseph; Bernier, Cynthia et al. (2016) The synthetic diazonamide DZ-2384 has distinct effects on microtubule curvature and dynamics without neurotoxicity. Sci Transl Med 8:365ra159
Ocal, Ozhan; Pashkov, Victor; Kollipara, Rahul K et al. (2015) A rapid in vivo screen for pancreatic ductal adenocarcinoma therapeutics. Dis Model Mech 8:1201-11
Kirane, Amanda; Ludwig, Kathleen F; Sorrelle, Noah et al. (2015) Warfarin Blocks Gas6-Mediated Axl Activation Required for Pancreatic Cancer Epithelial Plasticity and Metastasis. Cancer Res 75:3699-705