4.8 Article

Epigenetic silencing by SETDB1 suppresses tumour intrinsic immunogenicity

Journal

NATURE
Volume 595, Issue 7866, Pages 309-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41586-021-03520-4

Keywords

-

Funding

  1. Calico Life Sciences
  2. Gene Regulation Observatory at the Broad Institute of MIT and Harvard
  3. Damon-Runyon Cancer Research Foundation
  4. National Cancer Institute (NCI) Clinical Proteomic Tumor Analysis Consortium [NIH/NCI U24-CA210986, NIH/NCI U01 CA214125]
  5. Wong Family Award
  6. NCI/NIH Director's Fund [DP1CA216873]

Ask authors/readers for more resources

A CRISPR-Cas9 screen in mouse tumour models treated with immune checkpoint blockade identified SETDB1 as an epigenetic checkpoint protein that suppresses tumour-intrinsic immunogenicity. Loss of SETDB1 leads to derepression of immune-stimulating genes and triggers TE-specific cytotoxic T cell responses, suggesting it as a potential target for immunotherapy in cancer treatment.
A CRISPR-Cas9 screen of chromatin regulators in mouse tumour models treated with immune checkpoint blockade identifies SETDB1 as an epigenetic checkpoint protein that suppresses tumour-intrinsic immunogenicity. Epigenetic dysregulation is a defining feature of tumorigenesis that is implicated in immune escape(1,2). Here, to identify factors that modulate the immune sensitivity of cancer cells, we performed in vivo CRISPR-Cas9 screens targeting 936 chromatin regulators in mouse tumour models treated with immune checkpoint blockade. We identified the H3K9 methyltransferase SETDB1 and other members of the HUSH and KAP1 complexes as mediators of immune escape(3-5). We also found that amplification of SETDB1 (1q21.3) in human tumours is associated with immune exclusion and resistance to immune checkpoint blockade. SETDB1 represses broad domains, primarily within the open genome compartment. These domains are enriched for transposable elements (TEs) and immune clusters associated with segmental duplication events, a central mechanism of genome evolution(6). SETDB1 loss derepresses latent TE-derived regulatory elements, immunostimulatory genes, and TE-encoded retroviral antigens in these regions, and triggers TE-specific cytotoxic T cell responses in vivo. Our study establishes SETDB1 as an epigenetic checkpoint that suppresses tumour-intrinsic immunogenicity, and thus represents a candidate target for immunotherapy.

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