Abstract

Abstract Previous studies have concluded that the mechanism responsible for the antineoplastic properties of NSAIDs is cyclooxygenase (COX) independent, which suggests that more selective drugs can be developed for cancer chemoprevention by targeting such mechanisms. With this paradigm, we are synthesizing novel sulindac derivatives that have reduced COX inhibitory activity, but enhanced tumor cell growth inhibitory activity. A prototypic dimethylethyl amide derivative referred to as sulindac sulfide amide (SSA) was previously reported to lack COX-1 or COX-2 inhibitory activity, yet have improved potency to inhibit tumor cell growth in vitro. For example, SSA inhibited the growth of human HT-29 colon tumor cells with IC50 values of 1-2 µM compared with 70-90 µM for sulindac sulfide (SS), a non-selective COX inhibitor. SSA was also appreciably more effective than SS in inhibiting HT-29 colon tumor cell growth using a 3-dimensional in vitro tumor cell model. SSA induced apoptosis of HT-29 carcinoma cells, but normal human colonocytes were insensitive to treatment, suggesting an element of tumor selectivity. The mechanism of SSA induced apoptosis is associated with cGMP phosphodiesterase (PDE) inhibition, elevation of intracellular cGMP, and activation of protein kinase G, which also appears to be an important off-target effect of SS. SSA inhibited the growth of a variety of histologically diverse tumor cell lines, an effect that is consistent with studies showing the overexpression of PDE5 in various tumor types, including colorectal adenomas and carcinomas. SSA was well tolerated in the FCCC Min mouse model in which oral administration reduced colon tumor multiplicity by approximately 80% and tumor incidence by 50%. SSA also inhibited tumor growth in the human HT-29 colon mouse xenograft model by approximately 60%, while sulindac at its maximum tolerated dose was marginally effective. The antitumor activity of SSA in the HT-29 model was accompanied by decreased numbers of proliferating cells and increased numbers of apoptotic cells as determined by immunohistochemistry. Pharmacokinetic studies in mice bearing HT-29 xenografts revealed that plasma and tumor levels of SSA from a tolerated dose of SSA appreciably exceeded the in vitro IC50 value for growth inhibition. In contrast, sulindac generated plasma and tumor levels of SS that were appreciably less than the IC50 value for tumor cell growth inhibition. These results support the feasibility of chemically modifying sulindac to design out COX associated toxicities, while increasing anticancer efficacy. Newer sulindac derivatives with higher potency and target selectivity, as well as formulations to improve oral bioavailability are being developed for colon cancer chemoprevention. Funding provided by NIH/NCI grants CA131378 and CA148817. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1865. doi:10.1158/1538-7445.AM2011-1865

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