American Association for Cancer Research
Browse
TABLE_1.xls (5.5 kB)

TABLE 1 from Comparative Epigenetic Profiling Reveals Distinct Features of Mucosal Melanomas Associated with Immune Cell Infiltration and Their Clinical Implications

Download (5.5 kB)
dataset
posted on 2024-05-28, 14:20 authored by Jie Dai, Jia Jia, Fanshuang Zhang, Kaihua Liu, Yanfeng Xi, Pei Yuan, Lili Mao, Xue Bai, Xiaoting Wei, Bingning Wang, Jiangtao Li, Yang Xu, Ting Liu, Shuang Chang, Yang Shao, Jun Guo, Jianming Ying, Lu Si

Demographic and clinical characteristics of the patients with PMME and NEMM included in the study

Funding

MOST | National Natural Science Foundation of China (NSFC)

MOST | National Key Research and Development Program of China (NKPs)

Beijing Municipal Administration of Hospitals (北京市医院管理局)

Chinese American Medical Society (CAMS)

Vlove

北京市科学技术委员会 | Beijing Municipal Natural Science Foundation (北京市自然科学基金)

History

ARTICLE ABSTRACT

Mucosal melanoma exhibits limited responsiveness to anti-PD-1 therapy. However, a subgroup of mucosal melanomas, particularly those situated at specific anatomic sites like primary malignant melanoma of the esophagus (PMME), display remarkable sensitivity to anti-PD-1 treatment. The underlying mechanisms driving this superior response and the DNA methylation patterns in mucosal melanoma have not been thoroughly investigated. We collected tumor samples from 50 patients with mucosal melanoma, including 31 PMME and 19 non-esophageal mucosal melanoma (NEMM). Targeted bisulfite sequencing was conducted to characterize the DNA methylation landscape of mucosal melanoma and explore the epigenetic profiling differences between PMME and NEMM. Bulk RNA sequencing and multiplex immunofluorescence staining were performed to confirm the impact of methylation on gene expression and immune microenvironment. Our analysis revealed distinct epigenetic signatures that distinguish mucosal melanomas of different origins. Notably, PMME exhibited distinct epigenetic profiling characterized by a global hypermethylation alteration compared with NEMM. The prognostic model based on the methylation scores of a 7-DMR panel could effectively predict the overall survival of patients with PMME and potentially serve as a prognostic factor. PMME displayed a substantial enrichment of immune-activating cells in contrast to NEMM. Furthermore, we observed hypermethylation of the TERT promoter in PMME, which correlated with heightened CD8+ T-cell infiltration, and patients with hypermethylated TERT were likely to have improved responses to immunotherapy. Our results indicated that PMME shows a distinct methylation landscape compared with NEMM, and the epigenetic status of TERT might be used to estimate prognosis and direct anti-PD-1 treatment for mucosal melanoma. This study investigated the intricate epigenetic factor of mucosal melanomas contributed to the differential immune checkpoint inhibitor response, and found that PMME exhibited a global hypermethylation pattern and lower gene expression in comparison to NEMM. TERT hypermethylation may contribute to the favorable responses observed in patients with mucosal melanoma undergoing immunotherapy.