Abstract

Breast cancers are heterogeneous tissues comprised of multiple components, including tumor and microenvironment cells. Cancer‐associated fibroblasts (CAFs) are an important cell type in tumor‐associated stroma with multiple roles in tumor initiation and promotion. While the significance of CAFs is well recognized, the treatment of CAFs in clinical practice has not yet been established. Our previous studies have shown that the overexpression of phosphodiesterase (PDE) 5, a metallohydrolase which catalyzes the hydrolytic breakdown of cGMP, enhances the invasive potential of breast cancer cells and reduces survival in breast cancer patients. To extend these observations, we propose to assess the function of PDE5 in tumor microenvironment, to further highlight the potential benefit of targeting PDE5.Our results evidenced, for the first time, PDE5 mRNA and protein expression in CAFs isolated from biopsies of primary human breast tumors. PDE5 expression is increased in human breast cancer stroma compared with normal breast stroma and was correlated to a shorter overall survival in patients. Treatment with PDE5 inhibitors (i.e. sildenafil and tadalafil) significantly inhibited CAF proliferation, motility and invasiveness. To better examine the significance of PDE5 expression in stromal fibroblasts, PDE5 was stably integrated into mouse embryonic fibroblasts (MEFs) and cell proliferation, motility and invasion were investigated. PDE5 transformed MEFs towards a cancer‐associated fibroblast phenotype, as evidenced by increased expression of CAF markers along with enhanced proliferative, migratory and invasive capabilities. Coculture experiments showed that proliferation and motility of human breast cancer cells was increased by PDE5‐overexpressing MEFs compared to vector‐expressing MEFs, suggesting that stromal PDE5 contributes to heterotypic communications within breast cancer. An enriched production of the chemokine CXCL16 was revealed in PDE5‐expressing clones, as evidenced by cytokine array and confirmed by realtime PCR, suggesting a role for this molecule in mediating the tumor‐promoting effects of PDE5 expression in breast stroma. In line with these findings, exposure of CAFs to PDE5 inhibitors was associated with a reduced expression of CXCL16. More importantly, expression of CXCL16 in breast cancer stroma showed a strong correlation with PDE5 as well as with a poor patient survival.In conclusion, PDE5 is overexpressed in breast cancer stroma and enhances the tumor‐stimulatory activities of fibroblasts, highlighting this enzyme as a novel prognostic candidate and an attractive target for future therapy in breast cancers, having already addressed the delivery, stability, and toxicity issues of PDE5 inhibitors in other diseases.Support or Funding InformationMy First AIRC Grant (MFAG) #16899 to I. BaroneThis abstract is from the Experimental Biology 2019 Meeting. There is no full text article associated with this abstract published in The FASEB Journal.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call