Abstract

Abstract The incidence of melanoma is increasing faster than that of any other cancer, and predicted to double every 10-20 years. Surgery can be curative in Stage I, II, or III disease, but 75% of patients with deep primary lesions develop extensive recurrence or distant metastases and have dismal prognosis. In fact, there is no curative treatment for stage IV melanoma. Although novel targeted therapies, such as BRAF inhibitors and anti-CTLA4 antibodies are showing promising results in melanoma clinical trials, resistance to these agents and patient relapse rapidly ensue. Therapeutic resistance has been commonly attributed to functional redundancy between intimately hardwired cellular pathways responsible for tumor cell maintenance and survival. In order to avoid redundancy, we propose to directly inhibit the transcription of multiple genes required for the establishment or maintenance of tumors. First, we analyzed the expression of Bromodomain (BrD)-containing proteins, a family of epigenetic readers that bind acetylated lysine. BrDs are present in histone acetyl transferases (i.e. CBP/p300, PCAF, GCN5) and transcriptional regulators (i.e. BET family members: BRD2, 3, and 4). mRNA expression arrays showed several BrD-containing genes as upregulated in primary and metastatic melanoma cell lines compared to normal melanocytes. Analysis of available expression profiles also revealed higher levels of BRD2 and BRD4 in melanoma tissues relative to nevi or normal skin (P<0.001). Furthermore, immunohistochemistry staining of a melanoma tissue microarray confirmed overexpression of BRD4 protein in primary (P<0.001) and metastatic tumors (P<0.001) compared to nevi. BRD4 knockdown using siRNA or shRNA suppressed the proliferation and colony formation capacity of several metastatic melanoma cell lines. Moreover, melanoma cells stably infected with shBRD4 carrying lentivirus displayed reduced tumor growth in vivo compared with their counterpart infected with a non-silencing control. To identify cellular pathways modulated by BrD inhibition and to investigate the mechanism(s) that might mediate its anti-proliferative effects, we conducted a global transcriptome analysis (RNA sequencing) of 6 melanoma cell lines treated with specific BrD inhibitors or vehicle. Gene ontology analysis of these data revealed enrichment in genes controlling cell cycle, growth and proliferation. Accordingly, cell cycle analysis demonstrated a specific G1 phase arrest upon BRD4 silencing. In conclusion, we have demonstrated that expression of several BrD containing proteins is dysregulated in melanoma and that BRD4 represents a promising therapeutic target in these tumors. Our data provides a rationale for testing the efficacy of newly developed, specific and potent BET inhibitors against melanoma. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2185. doi:1538-7445.AM2012-2185

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