American Association for Cancer Research
Browse
00085472can150506-sup-145695_2_supp_0_ns5t1p.xlsx (379.41 kB)

Supplementary Table S5 from A Molecular Portrait of High-Grade Ductal Carcinoma In Situ

Download (379.41 kB)
dataset
posted on 2023-03-30, 23:09 authored by Martin C. Abba, Ting Gong, Yue Lu, Jaeho Lee, Yi Zhong, Ezequiel Lacunza, Matias Butti, Yoko Takata, Sally Gaddis, Jianjun Shen, Marcos R. Estecio, Aysegul A. Sahin, C. Marcelo Aldaz

Supplementary Table S5: Differentially methylated CpGi regions among normal and DCIS samples and their correlation with gene expression.

History

ARTICLE ABSTRACT

Ductal carcinoma in situ (DCIS) is a noninvasive precursor lesion to invasive breast carcinoma. We still have no understanding on why only some DCIS lesions evolve to invasive cancer whereas others appear not to do so during the life span of the patient. Here, we performed full exome (tumor vs. matching normal), transcriptome, and methylome analysis of 30 pure high-grade DCIS (HG-DCIS) and 10 normal breast epithelial samples. Sixty-two percent of HG-DCIS cases displayed mutations affecting cancer driver genes or potential drivers. Mutations were observed affecting PIK3CA (21% of cases), TP53 (17%), GATA3 (7%), MLL3 (7%) and single cases of mutations affecting CDH1, MAP2K4, TBX3, NF1, ATM, and ARID1A. Significantly, 83% of lesions displayed numerous large chromosomal copy number alterations, suggesting they might precede selection of cancer driver mutations. Integrated pathway-based modeling analysis of RNA-seq data allowed us to identify two DCIS subgroups (DCIS-C1 and DCIS-C2) based on their tumor-intrinsic subtypes, proliferative, immune scores, and in the activity of specific signaling pathways. The more aggressive DCIS-C1 (highly proliferative, basal-like, or ERBB2+) displayed signatures characteristic of activated Treg cells (CD4+/CD25+/FOXP3+) and CTLA4+/CD86+ complexes indicative of a tumor-associated immunosuppressive phenotype. Strikingly, all lesions showed evidence of TP53 pathway inactivation. Similarly, ncRNA and methylation profiles reproduce changes observed postinvasion. Among the most significant findings, we observed upregulation of lncRNA HOTAIR in DCIS-C1 lesions and hypermethylation of HOXA5 and SOX genes. We conclude that most HG-DCIS lesions, in spite of representing a preinvasive stage of tumor progression, displayed molecular profiles indistinguishable from invasive breast cancer. Cancer Res; 75(18); 3980–90. ©2015 AACR.