Targeting the lipid kinase PIKfyve upregulates surface expression of MHC class I to augment cancer immunotherapy

Author:

Bao Yi12,Qiao Yuanyuan123ORCID,Choi Jae Eun12,Zhang Yuping12,Mannan Rahul12ORCID,Cheng Caleb12,He Tongchen12,Zheng Yang12,Yu Jiali45,Gondal Mahnoor126,Cruz Gabriel1,Grove Sara45,Cao Xuhong12,Su Fengyun12,Wang Rui12ORCID,Chang Yu12,Kryczek Ilona45,Cieslik Marcin126,Green Michael D.578,Zou Weiping12345,Chinnaiyan Arul M.123910ORCID

Affiliation:

1. Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109

2. Department of Pathology, University of Michigan, Ann Arbor, MI 48109

3. Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109

4. Department of Surgery, University of Michigan, Ann Arbor, MI 48109

5. Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI 48109

6. Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109

7. Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109

8. Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48109

9. HHMI, University of Michigan, Ann Arbor, MI 48109

10. Department of Urology, University of Michigan, Ann Arbor, MI 48109

Abstract

Despite the remarkable clinical success of immunotherapies in a subset of cancer patients, many fail to respond to treatment and exhibit resistance. Here, we found that genetic or pharmacologic inhibition of the lipid kinase PIKfyve, a regulator of autophagic flux and lysosomal biogenesis, upregulated surface expression of major histocompatibility complex class I (MHC-I) in cancer cells via impairing autophagic flux, resulting in enhanced cancer cell killing mediated by CD8 + T cells. Genetic depletion or pharmacologic inhibition of PIKfyve elevated tumor-specific MHC-I surface expression, increased intratumoral functional CD8 + T cells, and slowed tumor progression in multiple syngeneic mouse models. Importantly, enhanced antitumor responses by Pikfyve -depletion were CD8 + T cell- and MHC-I-dependent, as CD8 + T cell depletion or B2m knockout rescued tumor growth. Furthermore, PIKfyve inhibition improved response to immune checkpoint blockade (ICB), adoptive cell therapy, and a therapeutic vaccine. High expression of PIKFYVE was also predictive of poor response to ICB and prognostic of poor survival in ICB-treated cohorts. Collectively, our findings show that targeting PIKfyve enhances immunotherapies by elevating surface expression of MHC-I in cancer cells, and PIKfyve inhibitors have potential as agents to increase immunotherapy response in cancer patients.

Funder

HHS | NIH | National Cancer Institute

Publisher

Proceedings of the National Academy of Sciences

Subject

Multidisciplinary

Cited by 1 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

全球学者库

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

"全球学者库"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,当前全球学者库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 © 2019-2023 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