4.7 Article

Human endogenous retroviruses as epigenetic therapeutic targets in TP53-mutated diffuse large B-cell lymphoma

Journal

Publisher

SPRINGERNATURE
DOI: 10.1038/s41392-023-01626-x

Keywords

-

Ask authors/readers for more resources

This study assessed the impact of TP53 mutation on diffuse large B-cell lymphoma (DLBCL) and its role in DLBCL treatment. The results showed that TP53 mutation is associated with poor prognosis in DLBCL patients treated with R-CHOP, but this effect can be mitigated by DR-CHOP treatment. TP53 mutation also leads to immune modulation repression and increased expression of SUV39H1 and H3K9 trimethylation, resulting in the inhibition of endogenous retroviruses (ERVs) and the formation of an immunosuppressive tumor microenvironment.
TP53 mutation (TP53(mut)) occurs in 10-20% of diffuse large B-cell lymphoma (DLBCL) cases and serves as an unfavorable biomarker of DLBCL progression. It confers resistance to immunochemotherapy, high-dose chemotherapy, autologous stem cell transplantation, and anti-CD19 chimeric antigen receptor T-cell therapy. Therapeutic targeting of TP53(mut) remains a significant challenge in DLBCL treatment. Here we assessed TP53(mut) in 667 patients with newly diagnosed DLBCL, including 576 patients treated with immunochemotherapy rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) and 91 patients with decitabine plus R-CHOP (DR-CHOP, NCT02951728 and NCT04025593). TP53(mut) independently predicted an inferior prognosis in R-CHOP-treated DLBCL, although this could be mitigated by DR-CHOP treatment. In TP53(mut) patients, multiple viral regulation pathways were repressed, resulting in the inhibition of immune modulation, as revealed by gene set enrichment analysis. TP53(mut) DLBCL exhibited increased methyltransferase SUV39H1 expression and H3K9 trimethylation (H3K9me3), contributing to repression of endogenous retroviruses (ERVs) and immunosuppressive tumor microenvironment. In TP53(mut) DLBCL cell lines, decitabine down-regulated SUV39H1, inhibited H3K9me3 occupancy on ERVs, and triggered ERV expression, thereby unleashing interferons program and CD4(+)T/CD8(+)T cell activation. Molecular silencing of SUV39H1 significantly abrogated decitabine-induced H3K9me3 inhibition and ERV expression. In TP53(mut) patient-derived xenograft models and TP53(mut) patients, the anti-tumor effect was improved upon the use of combined treatment of decitabine and doxorubicin via SUV39H1-H3K9me3-ERVs axis. Collectively, our findings highlight an ERV regulatory circuitry in TP53(mut) DLBCL and the crucial roles ERVs for epigenetically reprogramming tumor microenvironment for treating TP53(mut)-driven cancers.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available