Skip to main content

SETDB1 Restrains Endogenous Retrovirus Expression and Antitumor Immunity during Radiotherapy.

Publication ,  Journal Article
Pan, D; Bao, X; Hu, M; Jiao, M; Li, F; Li, C-Y
Published in: Cancer Res
August 3, 2022

UNLABELLED: The type I interferon response plays a pivotal role in promoting antitumor immune activity in response to radiotherapy. The identification of approaches to boost the radiation-induced type I interferon response could help improve the efficacy of radiotherapy. Here we show that the histone methyltransferase SETDB1 is a potent suppressor of radiation-induced endogenous retrovirus expression. SETDB1 inhibition significantly enhanced the efficacy of radiotherapy by promoting radiation-induced viral mimicry to upregulate type I interferons. SETDB1 expression correlated with radiotherapy efficacy in human non-small cell carcinoma and melanoma patients. In a murine tumor model, genetic deletion of Setdb1 significantly enhanced radiotherapy efficacy, and Setdb1-deficient tumors had enhanced intratumoral lymphocyte infiltration, an observation confirmed in human cancer samples. Setdb1 deficiency led to increased basal and radiation-induced endogenous retrovirus (ERV) expression, enhanced MDA5/MAVS signaling, and upregulated type I interferons, which were essential for SETDB1 deficiency-induced radiosensitization. Taken together, these data suggest that inhibition of SETDB1 is a promising approach to enhance cancer radiotherapy efficacy by promoting radiation-induced viral mimicry and antitumor immunity through ERV induction. SIGNIFICANCE: The identification of the SETDB1-mediated suppression of radiotherapy-induced viral mimicry reveals SETDB1 inhibition as a potential approach to sensitize tumors to radiotherapy by enhancing the type I interferon response.

Duke Scholars

Altmetric Attention Stats
Dimensions Citation Stats

Published In

Cancer Res

DOI

EISSN

1538-7445

Publication Date

August 3, 2022

Volume

82

Issue

15

Start / End Page

2748 / 2760

Location

United States

Related Subject Headings

  • Signal Transduction
  • Oncology & Carcinogenesis
  • Mice
  • Melanoma
  • Interferon Type I
  • Humans
  • Histone-Lysine N-Methyltransferase
  • Endogenous Retroviruses
  • Animals
  • 3211 Oncology and carcinogenesis
 

Citation

APA
Chicago
ICMJE
MLA
NLM
Pan, D., Bao, X., Hu, M., Jiao, M., Li, F., & Li, C.-Y. (2022). SETDB1 Restrains Endogenous Retrovirus Expression and Antitumor Immunity during Radiotherapy. Cancer Res, 82(15), 2748–2760. https://doi.org/10.1158/0008-5472.CAN-21-3523
Pan, Dong, Xuhui Bao, Mengjie Hu, Meng Jiao, Fang Li, and Chuan-Yuan Li. “SETDB1 Restrains Endogenous Retrovirus Expression and Antitumor Immunity during Radiotherapy.Cancer Res 82, no. 15 (August 3, 2022): 2748–60. https://doi.org/10.1158/0008-5472.CAN-21-3523.
Pan D, Bao X, Hu M, Jiao M, Li F, Li C-Y. SETDB1 Restrains Endogenous Retrovirus Expression and Antitumor Immunity during Radiotherapy. Cancer Res. 2022 Aug 3;82(15):2748–60.
Pan, Dong, et al. “SETDB1 Restrains Endogenous Retrovirus Expression and Antitumor Immunity during Radiotherapy.Cancer Res, vol. 82, no. 15, Aug. 2022, pp. 2748–60. Pubmed, doi:10.1158/0008-5472.CAN-21-3523.
Pan D, Bao X, Hu M, Jiao M, Li F, Li C-Y. SETDB1 Restrains Endogenous Retrovirus Expression and Antitumor Immunity during Radiotherapy. Cancer Res. 2022 Aug 3;82(15):2748–2760.

Published In

Cancer Res

DOI

EISSN

1538-7445

Publication Date

August 3, 2022

Volume

82

Issue

15

Start / End Page

2748 / 2760

Location

United States

Related Subject Headings

  • Signal Transduction
  • Oncology & Carcinogenesis
  • Mice
  • Melanoma
  • Interferon Type I
  • Humans
  • Histone-Lysine N-Methyltransferase
  • Endogenous Retroviruses
  • Animals
  • 3211 Oncology and carcinogenesis