Targeting KDM2A Enhances T-cell Infiltration in NSD1-Deficient Head and Neck Squamous Cell Carcinoma

Author:

Chen Chen12ORCID,Shin June Ho12ORCID,Fang Zhuoqing3ORCID,Brennan Kevin24ORCID,Horowitz Nina B.12ORCID,Pfaff Kathleen L.56ORCID,Welsh Emma L.56ORCID,Rodig Scott J.67ORCID,Gevaert Olivier24ORCID,Gozani Or28ORCID,Uppaluri Ravindra79ORCID,Sunwoo John B.12ORCID

Affiliation:

1. 1Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, Stanford, California.

2. 2Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.

3. 3Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California.

4. 4Department of Medicine (Biomedical Informatics) and Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, California.

5. 5Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.

6. 6Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.

7. 7Dana-Farber Cancer Institute, Boston, Massachusetts.

8. 8Department of Biology, Stanford University, Stanford, California.

9. 9Division of Otolaryngology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.

Abstract

Abstract In head and neck squamous cell carcinoma (HNSCC), a significant proportion of tumors have inactivating mutations in the histone methyltransferase NSD1. In these tumors, NSD1 inactivation is a driver of T-cell exclusion from the tumor microenvironment (TME). A better understanding of the NSD1-mediated mechanism regulating infiltration of T cells into the TME could help identify approaches to overcome immunosuppression. Here, we demonstrated that NSD1 inactivation results in lower levels of H3K36 dimethylation and higher levels of H3K27 trimethylation, the latter being a known repressive histone mark enriched on the promoters of key T-cell chemokines CXCL9 and CXCL10. HNSCC with NSD1 mutations had lower levels of these chemokines and lacked responses to PD-1 immune checkpoint blockade. Inhibition of KDM2A, the primary lysine demethylase that is selective for H3K36, reversed the altered histone marks induced by NSD1 loss and restored T-cell infiltration into the TME. Importantly, KDM2A suppression decreased growth of NSD1-deficient tumors in immunocompetent, but not in immunodeficient, mice. Together, these data indicate that KDM2A is an immunotherapeutic target for overcoming immune exclusion in HNSCC. Significance: The altered epigenetic landscape of NSD1-deficient tumors confers sensitivity to inhibition of the histone-modifying enzyme KDM2A as an immunotherapeutic strategy to stimulate T-cell infiltration and suppress tumor growth.

Funder

National Institute of Dental and Craniofacial Research

Publisher

American Association for Cancer Research (AACR)

Subject

Cancer Research,Oncology

Cited by 2 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

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