Low level of ARID1A contributes to adaptive immune resistance and sensitizes triple‐negative breast cancer to immune checkpoint inhibitors

Author:

Chen Xin‐Yu12,Li Bin12,Wang Ye12,Jin Juan12,Yang Yu3,Huang Lei‐Huan3,Yang Meng‐Di12,Zhang Jian12,Wang Bi‐Yun12ORCID,Shao Zhi‐Ming245,Ni Ting3,Huang Sheng‐Lin26,Hu Xi‐Chun12ORCID,Tao Zhong‐Hua12ORCID

Affiliation:

1. Department of Breast and Urologic Medical Oncology Fudan University Shanghai Cancer Center Shanghai P. R. China

2. Department of Oncology Shanghai Medical College Fudan University Shanghai P. R. China

3. State Key Laboratory of Genetic Engineering Collaborative Innovation Center of Genetics and Development Human Phenome Institute School of Life Sciences Fudan University Shanghai P. R. China

4. Key Laboratory of Breast Cancer in Shanghai Department of Breast Surgery Fudan University Shanghai Cancer Center Shanghai P. R. China

5. Precision Cancer Medicine Center Fudan University Shanghai Cancer Center Shanghai P. R. China

6. Shanghai Key Laboratory of Medical Epigenetics International Co‐laboratory of Medical Epigenetics and Metabolism Institutes of Biomedical Sciences Shanghai Medical College Fudan University Shanghai P. R. China

Abstract

AbstractBackgroundImmune checkpoint inhibitors (ICIs) shed new light on triple‐negative breast cancer (TNBC), but only a minority of patients demonstrate response. Therefore, adaptive immune resistance (AIR) needs to be further defined to guide the development of ICI regimens.MethodsDatabases, including The Cancer Genome Atlas, Gene Ontology Resource, University of California Santa Cruz Genome Browser, and Pubmed, were used to screen epigenetic modulators, regulators for CD8+ T cells, and transcriptional regulators of programmed cell death‐ligand 1 (PD‐L1). Human peripheral blood mononuclear cell (Hu‐PBMC) reconstruction mice were adopted for xenograft transplantation. Tumor specimens from a TNBC cohort and the clinical trial CTR20191353 were retrospectively analyzed. RNA‐sequencing, Western blotting, qPCR and immunohistochemistry were used to assess gene expression. Coculture assays were performed to evaluate the regulation of TNBC cells on T cells. Chromatin immunoprecipitation and transposase‐accessible chromatin sequencing were used to determine chromatin‐binding and accessibility.ResultsThe epigenetic modulator AT‐rich interaction domain 1A (ARID1A) gene demonstrated the highest expression association with AIR relative to other epigenetic modulators in TNBC patients. Low ARID1A expression in TNBC, causing an immunosuppressive microenvironment, promoted AIR and inhibited CD8+ T cell infiltration and activity through upregulating PD‐L1. However, ARID1A did not directly regulate PD‐L1 expression. We found that ARID1A directly bound the promoter of nucleophosmin 1 (NPM1) and that low ARID1A expression increased NPM1 chromatin accessibility as well as gene expression, further activating PD‐L1 transcription. In Hu‐PBMC mice, atezolizumab demonstrated the potential to reverse ARID1A deficiency‐induced AIR in TNBC by reducing tumor malignancy and activating anti‐tumor immunity. In CTR20191353, ARID1A‐low patients derived more benefit from pucotenlimab compared to ARID1A‐high patients.ConclusionsIn AIR epigenetics, low ARID1A expression in TNBC contributed to AIR via the ARID1A/NPM1/PD‐L1 axis, leading to poor outcome but sensitivity to ICI treatment.

Funder

National Science and Technology Major Project

Publisher

Wiley

Subject

Cancer Research,Oncology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3