American Association for Cancer Research
Browse
00085472can142860-sup-138500_1_supp_2878147_nkdy13_eps_zip.zip (414.68 kB)

Supplemental Figure S2 from KRAS Genomic Status Predicts the Sensitivity of Ovarian Cancer Cells to Decitabine

Download (414.68 kB)
software
posted on 2023-03-30, 23:09 authored by Michelle L. Stewart, Pablo Tamayo, Andrew J. Wilson, Stephanie Wang, Yun Min Chang, Jong W. Kim, Dineo Khabele, Alykhan F. Shamji, Stuart L. Schreiber

A, cell viability after treatment with decitabine (AUC, x-axis) was plotted against the doubling time for each cell line (y-axis) as measured over nine days in 384-well plates at each cell line's optimized density. B, cell viability was measured after treatment with decitabine for nine days in four KRAS-mutant pancreatic cell lines. Data are representative of two independent experiments (14 replicates, mean {plus minus} SD). C, cell viability was measured after treatment with decitabine for nine days in KRAS-mutant (purple) or KRAS¬¬¬-wild type colon (gray) cancer cell lines. Data are representative of two independent experiments. Data are representative of two independent experiments (14 replicates, mean {plus minus} SD).

History

ARTICLE ABSTRACT

Decitabine, a cancer therapeutic that inhibits DNA methylation, produces variable antitumor response rates in patients with solid tumors that might be leveraged clinically with identification of a predictive biomarker. In this study, we profiled the response of human ovarian, melanoma, and breast cancer cells treated with decitabine, finding that RAS/MEK/ERK pathway activation and DNMT1 expression correlated with cytotoxic activity. Further, we showed that KRAS genomic status predicted decitabine sensitivity in low-grade and high-grade serous ovarian cancer cells. Pretreatment with decitabine decreased the cytotoxic activity of MEK inhibitors in KRAS-mutant ovarian cancer cells, with reciprocal downregulation of DNMT1 and MEK/ERK phosphorylation. In parallel with these responses, decitabine also upregulated the proapoptotic BCL-2 family member BNIP3, which is known to be regulated by MEK and ERK, and heightened the activity of proapoptotic small-molecule navitoclax, a BCL-2 family inhibitor. In a xenograft model of KRAS-mutant ovarian cancer, combining decitabine and navitoclax heightened antitumor activity beyond administration of either compound alone. Our results define the RAS/MEK/DNMT1 pathway as a determinant of sensitivity to DNA methyltransferase inhibition, specifically implicating KRAS status as a biomarker of drug response in ovarian cancer. Cancer Res; 75(14); 2897–906. ©2015 AACR.