4.8 Article

CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy

Journal

NATURE
Volume 569, Issue 7755, Pages 270-+

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/s41586-019-1170-y

Keywords

-

Funding

  1. NIH/NCI R01 grants [CA217648, CA123088, CA099985, CA193136, CA152470]
  2. NIH through the University of Michigan Rogel Cancer Center Support Grant [CA46592]
  3. NIH/NCI [CA189623, CA205426]
  4. Pershing Square Sohn Cancer Research
  5. PaineWebber Chair
  6. STARR Cancer Consortium
  7. NCI R35 [CA232097]
  8. NIH/NCI Cancer Center Support Grant [P30 CA008748]

Ask authors/readers for more resources

Cancer immunotherapy restores or enhances the effector function of CD8(+) T cells in the tumour microenvironment(1,2). CD8(+) T cells activated by cancer immunotherapy clear tumours mainly by inducing cell death through perforin-granzyme and Fas-Fas ligand pathways(3,4). Ferroptosis is a form of cell death that differs from apoptosis and results from iron-dependent accumulation of lipid peroxide(5,6). Although it has been investigated in vitro(7,8), there is emerging evidence that ferroptosis might be implicated in a variety of pathological scenarios(9,10). It is unclear whether, and how, ferroptosis is involved in T cell immunity and cancer immunotherapy. Here we show that immunotherapy-activated CD8(+) T cells enhance ferroptosis-specific lipid peroxidation in tumour cells, and that increased ferroptosis contributes to the antitumour efficacy of immunotherapy. Mechanistically, interferon gamma (IFN gamma) released from CD8(+) T cells downregulates the expression of SLC3A2 and SLC7A11, two subunits of the glutamate-cystine antiporter system x(c)(-), impairs the uptake of cystine by tumour cells, and as a consequence, promotes tumour cell lipid peroxidation and ferroptosis. In mouse models, depletion of cystine or cysteine by cyst(e)inase (an engineered enzyme that degrades both cystine and cysteine) in combination with checkpoint blockade synergistically enhanced T cell-mediated anti-tumour immunity and induced ferroptosis in tumour cells. Expression of system x(c)(- )was negatively associated, in cancer patients, with CD8(+) T cell signature, IFN gamma expression, and patient outcome. Analyses of human transcriptomes before and during nivolumab therapy revealed that clinical benefits correlate with reduced expression of SLC3A2 and increased IFN gamma and CD8. Thus, T cell-promoted tumour ferroptosis is an anti-tumour mechanism, and targeting this pathway in combination with checkpoint blockade is a potential therapeutic approach.

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