Cell intrinsic role of COX-2 in pancreatic cancer development

R Hill, Y Li, LM Tran, S Dry, JH Calvopina… - Molecular cancer …, 2012 - AACR
R Hill, Y Li, LM Tran, S Dry, JH Calvopina, A Garcia, C Kim, Y Wang, TR Donahue…
Molecular cancer therapeutics, 2012AACR
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 …
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-rasG12D/+; Ptenlox/+ 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. Mol Cancer Ther; 11(10); 2127–37. ©2012 AACR.
AACR