4.8 Article

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

Journal

CELL RESEARCH
Volume 30, Issue 11, Pages 966-979

Publisher

SPRINGERNATURE
DOI: 10.1038/s41422-020-00395-4

Keywords

-

Categories

Funding

  1. National Natural Science Foundation of China [31830022, 81621001, 31991171, 81830002, 81773248]
  2. Chinese Ministry of Science and Technology [2019YFA0508500]

Ask authors/readers for more resources

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(2+)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(2+)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(2+)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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available