Inhibiting Type I Arginine Methyltransferase Activity Promotes T Cell–Mediated Antitumor Immune Responses

Author:

Fedoriw Andrew1,Shi Leilei2,O'Brien Shane1ORCID,Smitheman Kimberly N.1,Wang Yunfei2ORCID,Hou Jiakai3,Sherk Christian1,Rajapurkar Satyajit1,Laraio Jenny1,Williams Leila J.2ORCID,Xu Chunyu3,Han Guangchun4,Feng Qin3,Bedford Mark T.5ORCID,Wang Linghua4ORCID,Barbash Olena1,Kruger Ryan G.1,Hwu Patrick2,Mohammad Helai P.1,Peng Weiyi3ORCID

Affiliation:

1. 1Tumor Cell Targeting Research Unit, GlaxoSmithKline, Collegeville, Pennsylvania.

2. 2Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.

3. 3Department of Biology and Biochemistry, University of Houston, Houston, Texas.

4. 4Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.

5. 5Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas.

Abstract

Abstract Protein arginine methyltransferases (PRMT) are a widely expressed class of enzymes responsible for catalyzing arginine methylation on numerous protein substrates. Among them, type I PRMTs are responsible for generating asymmetric dimethylarginine. By controlling multiple basic cellular processes, such as DNA damage responses, transcriptional regulation, and mRNA splicing, type I PRMTs contribute to cancer initiation and progression. A type I PRMT inhibitor, GSK3368715, has been developed and has entered clinical trials for solid and hematologic malignancies. Although type I PRMTs have been reported to play roles in modulating immune cell function, the immunologic role of tumor-intrinsic pathways controlled by type I PRMTs remains uncharacterized. Here, our The Cancer Genome Atlas dataset analysis revealed that expression of type I PRMTs associated with poor clinical response and decreased immune infiltration in patients with melanoma. In cancer cell lines, inhibition of type I PRMTs induced an IFN gene signature, amplified responses to IFN and innate immune signaling, and decreased expression of the immunosuppressive cytokine VEGF. In immunocompetent mouse tumor models, including a model of T-cell exclusion that represents a common mechanism of anti–programmed cell death protein 1 (PD-1) resistance in humans, type I PRMT inhibition increased T-cell infiltration, produced durable responses dependent on CD8+ T cells, and enhanced efficacy of anti–PD-1 therapy. These data indicate that type I PRMT inhibition exhibits immunomodulatory properties and synergizes with immune checkpoint blockade (ICB) to induce durable antitumor responses in a T cell–dependent manner, suggesting that type I PRMT inhibition can potentiate an antitumor immunity in refractory settings.

Funder

NCI

Cancer Prevention and Research Institute of Texas

NIH

Publisher

American Association for Cancer Research (AACR)

Subject

Cancer Research,Immunology

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