Cancer Therapy : Preclinical Vemurafenib SynergizeswithNutlin-3 toDeplete Survivin and Suppresses Melanoma Viability and Tumor Growth

Zhenyu Ji,Raj Kumar,Michael Taylor,Anpuchchelvi Rajadurai, Alexander Marzuka-Alcal, Y. Erin Chen, Ching-Ni Jenny Njauw,Keith Flaherty,Hensin Tsao

semanticscholar(2013)

引用 0|浏览0
暂无评分
摘要
Purpose: For patients with advanced melanoma, primary and secondary resistance to selective BRAF inhibition remains one of the most critically compelling challenges. One rationale argues that novel biologically informed strategies are needed to maximally cripple melanoma cells up front before compensatory mechanisms emerge. As p53 is uncommonly mutated in melanoma, restoration of its function represents an attractive adjunct to selective BRAF inhibition. Experimental Design: Thirty-seven BRAF(V600E)-mutated melanoma lines were subjected to synergy studies in vitro using a combination of vemurafenib and nutlin-3 (Nt-3). In addition, cellular responses and in vivo efficacy were also determined. We also analyzed changes in the levels of canonical apoptotic/survival factors in response to vemurafenib. Results: Dual targeting of BRAF(V600E) and Hdm2 with vemurafenib and Nt-3, respectively, synergistically induced apoptosis and suppressedmelanoma viability in vitro and tumor growth in vivo. Suppression of p53 in melanoma cells abrogated Nt-30s effects fully and vemurafenib’s effects partially. A survey of canonical survival factors revealed that both vemurafenib and Nt-3 independently attenuated levels of the antiapoptotic protein, survivin. Genetic depletion of survivin reproduces the cytotoxic effects of the combination strategy. Conclusion: These results show preclinical feasibility for overcoming primary vemurafenib resistance by restoring p53 function. Moreover, it identifies survivin as one downstream mediator of the observed synergism and a potential secondary target. Clin Cancer Res; 19(16); 4383–91. 2013 AACR.
更多
查看译文
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要