Cell intrinsic role of COX-2 in pancreatic cancer development

Mol Cancer Ther. 2012 Oct;11(10):2127-37. doi: 10.1158/1535-7163.MCT-12-0342. Epub 2012 Jul 10.

Abstract

COX-2 is upregulated in pancreatic ductal adenocarcinomas (PDAC). However, how COX-2 promotes PDAC development is unclear. While previous studies have evaluated the efficacy of COX-2 inhibition via the use of nonsteroidal anti-inflammatory drugs (NSAID) or the COX-2 inhibitor celecoxib in PDAC models, none have addressed the cell intrinsic versus microenvironment roles of COX-2 in modulating PDAC initiation and progression. We tested the cell intrinsic role of COX-2 in PDAC progression using both loss-of-function and gain-of-function approaches. Cox-2 deletion in Pdx1+ pancreatic progenitor cells significantly delays the development of PDAC in mice with K-ras activation and Pten haploinsufficiency. Conversely, COX-2 overexpression promotes early onset and progression of PDAC in the K-ras mouse model. Loss of PTEN function is a critical factor in determining lethal PDAC onset and overall survival. Mechanistically, COX-2 overexpression increases p-AKT levels in the precursor lesions of Pdx1(+); K-ras(G12D)(/+); Pten(lox)(/+) mice in the absence of Pten LOH. In contrast, Cox-2 deletion in the same setting diminishes p-AKT levels and delays cancer progression. These data suggest an important cell intrinsic role for COX-2 in tumor initiation and progression through activation of the PI3K/AKT pathway. PDAC that is independent of intrinsic COX-2 expression eventually develops with decreased FKBP5 and increased GRP78 expression, two alternate pathways leading to AKT activation. Together, these results support a cell intrinsic role for COX-2 in PDAC development and suggest that while anti-COX-2 therapy may delay the development and progression of PDAC, mechanisms known to increase chemoresistance through AKT activation must also be overcome.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Carcinoma, Pancreatic Ductal / enzymology
  • Carcinoma, Pancreatic Ductal / pathology
  • Celecoxib
  • Cell Membrane / drug effects
  • Cell Membrane / metabolism
  • Cyclooxygenase 2 / metabolism*
  • Disease Models, Animal
  • Disease Progression
  • Endoplasmic Reticulum Chaperone BiP
  • Enzyme Activation / drug effects
  • Feedback, Physiological / drug effects
  • Gene Deletion
  • Gene Targeting
  • Heat-Shock Proteins / metabolism
  • Homeodomain Proteins
  • Integrases / metabolism
  • Life Expectancy
  • Mice
  • PTEN Phosphohydrolase / metabolism
  • Pancreatic Neoplasms / enzymology*
  • Pancreatic Neoplasms / pathology*
  • Precancerous Conditions / enzymology*
  • Precancerous Conditions / pathology*
  • Proto-Oncogene Proteins c-akt / metabolism
  • Proto-Oncogene Proteins p21(ras) / metabolism
  • Pyrazoles / chemistry
  • Pyrazoles / pharmacology
  • Sulfonamides / chemistry
  • Sulfonamides / pharmacology
  • Survival Analysis
  • Tacrolimus Binding Proteins / metabolism
  • Trans-Activators
  • Up-Regulation / drug effects

Substances

  • Endoplasmic Reticulum Chaperone BiP
  • Heat-Shock Proteins
  • Homeodomain Proteins
  • Hspa5 protein, mouse
  • Pyrazoles
  • Sulfonamides
  • Trans-Activators
  • pancreatic and duodenal homeobox 1 protein
  • Ptgs2 protein, mouse
  • Cyclooxygenase 2
  • Proto-Oncogene Proteins c-akt
  • Cre recombinase
  • Integrases
  • PTEN Phosphohydrolase
  • Hras protein, mouse
  • Proto-Oncogene Proteins p21(ras)
  • Tacrolimus Binding Proteins
  • tacrolimus binding protein 5
  • Celecoxib