Manganese is critical for antitumor immune responses via cGAS-STING and improves the efficacy of clinical immunotherapy

CD8 T cell-mediated cancer clearance is often suppressed by the interaction between inhibitory molecules like PD-1 and PD-L1, an interaction acts like brakes to prevent T cell overreaction under normal conditions but is exploited by tumor cells to escape the immune surveillance. Immune checkpoint in...

Full description

Saved in:
Bibliographic Details
Published in:Cell research 2020-11, Vol.30 (11), p.966-979
Main Authors: Lv, Mengze, Chen, Meixia, Zhang, Rui, Zhang, Wen, Wang, Chenguang, Zhang, Yan, Wei, Xiaoming, Guan, Yukun, Liu, Jiejie, Feng, Kaichao, Jing, Miao, Wang, Xurui, Liu, Yun-Cai, Mei, Qian, Han, Weidong, Jiang, Zhengfan
Format: Article
Language:eng
Subjects:
Online Access:Get full text
Tags: Add Tag
No Tags, Be the first to tag this record!
Description
Summary:CD8 T cell-mediated cancer clearance is often suppressed by the interaction between inhibitory molecules like PD-1 and PD-L1, an interaction acts like brakes to prevent T cell overreaction under normal conditions but is exploited by tumor cells to escape the immune surveillance. Immune checkpoint inhibitors have revolutionized cancer therapeutics by removing such brakes. Unfortunately, only a minority of cancer patients respond to immunotherapies presumably due to inadequate immunity. Antitumor immunity depends on the activation of the cGAS-STING pathway, as STING-deficient mice fail to stimulate tumor-infiltrating dendritic cells (DCs) to activate CD8 T cells. STING agonists also enhance natural killer (NK) cells to mediate the clearance of CD8 T cell-resistant tumors. Therefore STING agonists have been intensively sought after. We previously discovered that manganese (Mn) is indispensable for the host defense against cytosolic dsDNA by activating cGAS-STING. Here we report that Mn is also essential in innate immune sensing of tumors and enhances adaptive immune responses against tumors. Mn-insufficient mice had significantly enhanced tumor growth and metastasis, with greatly reduced tumor-infiltrating CD8 T cells. Mechanically, Mn promoted DC and macrophage maturation and tumor-specific antigen presentation, augmented CD8 T cell differentiation, activation and NK cell activation, and increased memory CD8 T cells. Combining Mn with immune checkpoint inhibition synergistically boosted antitumor efficacies and reduced the anti-PD-1 antibody dosage required in mice. Importantly, a completed phase 1 clinical trial with the combined regimen of Mn and anti-PD-1 antibody showed promising efficacy, exhibiting type I IFN induction, manageable safety and revived responses to immunotherapy in most patients with advanced metastatic solid tumors. We propose that this combination strategy warrants further clinical translation.
ISSN:1001-0602
1748-7838