Abstract

Background: Epigenetic regulation, including DNA methylation, plays a major role in shaping the identity and function of immune cells. Innate and adaptive immune cells recruited into tumor tissues contribute to the formation of the tumor immune microenvironment (TIME), which is closely involved in tumor progression in breast cancer (BC). However, the specific methylation signatures of immune cells have not been thoroughly investigated yet. Additionally, it remains unknown whether immune cells-specific methylation signatures can identify subgroups and stratify the prognosis of BC patients.Methods: DNA methylation profiles of six immune cell types from eight datasets downloaded from the Gene Expression Omnibus were collected to identify immune cell-specific hypermethylation signatures (IC-SHMSs). Univariate and multivariate cox regression analyses were performed using BC data obtained from The Cancer Genome Atlas to identify the prognostic value of these IC-SHMSs. An unsupervised clustering analysis of the IC-SHMSs with prognostic value was performed to categorize BC patients into subgroups. Multiple Cox proportional hazard models were constructed to explore the role of IC-SHMSs and their relationship to clinical characteristics in the risk stratification of BC patients. Integrated discrimination improvement (IDI) was performed to determine whether the improvement of IC-SHMSs on clinical characteristics in risk stratification was statistically significant.Results: A total of 655 IC-SHMSs of six immune cell types were identified. Thirty of them had prognostic value, and 10 showed independent prognostic value. Four subgroups of BC patients, which showed significant heterogeneity in terms of survival prognosis and immune landscape, were identified. The model incorporating nine IC-SHMSs showed similar survival prediction accuracy as the clinical model incorporating age and TNM stage [3-year area under the curve (AUC): 0.793 vs. 0.785; 5-year AUC: 0.735 vs. 0.761]. Adding the IC-SHMSs to the clinical model significantly improved its prediction accuracy in risk stratification (3-year AUC: 0.897; 5-year AUC: 0.856). The results of IDI validated the statistical significance of the improvement (p < 0.05).Conclusions: Our study suggests that IC-SHMSs may serve as signatures of classification and risk stratification in BC. Our findings provide new insights into epigenetic signatures, which may help improve subgroup identification, risk stratification, and treatment management.

Highlights

  • Breast cancer (BC) is the most common cancer among women worldwide

  • The Cancer Genome Atlas (TCGA) level 3 gene expression data normalized by fragments per kilobase of exon per million reads mapped (FPKM), DNA methylation data, and somatic mutation data from BC patients, which were downloaded from the National Cancer Institute’s Genomic Data Commons Portal (GDC; https://portal.gdc.cancer.gov/); matched survival and clinical information were obtained from the University of Santa Cruz (UCSC) Xena database

  • Principal component analysis (PCA) analysis of immune cells was performed based on the methylation profiles of these 655 IC-SHMSs

Read more

Summary

Introduction

Breast cancer (BC) is the most common cancer among women worldwide. Despite significant advances in locoregional therapies, endocrine therapies, chemotherapy, and molecular targeted therapy, BC remains the second leading cause of cancerrelated deaths among women [1, 2]. Tumors can induce local immune dysregulation by suppressing innate and adaptive immune responses in BC [3]. The tumor immune microenvironment (TIME) is an important part of the tumor microenvironment It is highly heterogeneous and plays an important role in tumor progression and disease prognosis in various cancers [4]. Innate and adaptive immune cells recruited into tumor tissues contribute to the formation of the tumor immune microenvironment (TIME), which is closely involved in tumor progression in breast cancer (BC). The specific methylation signatures of immune cells have not been thoroughly investigated yet. It remains unknown whether immune cells-specific methylation signatures can identify subgroups and stratify the prognosis of BC patients

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.