Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockade

Background BANF1 is well known as a natural opponent of cyclic GMP-AMP synthase (cGAS) activity on genomic self-DNA. However, the roles of BANF1 in tumor immunity remain unclear. Here, we investigate the possible impact of BANF1 on antitumor immunity and response to immunotherapy.Methods The Cancer...

Full description

Bibliographic Details
Main Authors: Fei Wang, Yang Li, Jinming Yu, Dawei Chen, Fei Wu, Meng Wu, Minglei Wang, Yiheng Huang, Minxin Chen, Weiyan Wang, Tao Zhong, Xiaozheng Chen
Format: Article
Language:English
Published: BMJ Publishing Group 2023-08-01
Series:Journal for ImmunoTherapy of Cancer
Online Access:https://jitc.bmj.com/content/11/8/e007035.full
_version_ 1797729193826451456
author Fei Wang
Yang Li
Jinming Yu
Dawei Chen
Fei Wu
Meng Wu
Minglei Wang
Yiheng Huang
Minxin Chen
Weiyan Wang
Tao Zhong
Xiaozheng Chen
author_facet Fei Wang
Yang Li
Jinming Yu
Dawei Chen
Fei Wu
Meng Wu
Minglei Wang
Yiheng Huang
Minxin Chen
Weiyan Wang
Tao Zhong
Xiaozheng Chen
author_sort Fei Wang
collection DOAJ
description Background BANF1 is well known as a natural opponent of cyclic GMP-AMP synthase (cGAS) activity on genomic self-DNA. However, the roles of BANF1 in tumor immunity remain unclear. Here, we investigate the possible impact of BANF1 on antitumor immunity and response to immunotherapy.Methods The Cancer Genome Atlas public data were analyzed to evaluate the relevance of the expression of BANF1, patients’ survival and immune cell infiltration. We monitored tumor growth and explored the antitumor efficacy of targeting tumor-intrinsic BANF1 in combination with anti-programmed cell death protein-1 (PD-1) in MC38 or B16F10 tumor models in both immunocompetent and immunodeficient mice. Flow cytometry, immunofluorescence and T cells depletion experiments were used to validate the role of BANF1 in tumor immune microenvironment reprogramming. RNA sequencing was then used to interrogate the mechanisms how BANF1 regulated antitumor immunity.Results We show that upregulated expression of BANF1 in tumor tissues is significantly associated with poor survival and is negatively correlated with immune cell infiltration. Deficiency of BANF1 in tumor cells markedly antagonizes tumor growth in immunocompetent but not immunocompromised mice, and enhances the response to immunotherapy in murine models of melanoma and colon cancer. In the immunotherapy clinical cohort, patients with high BANF1 expression had a worse prognosis. Mechanistically, BANF1 knockout activates antitumor immune responses mediated by cGAS-synthase-stimulator of interferon genes (cGAS-STING) pathway, resulting in an immune-activating tumor microenvironment including increased CD8+ T cell infiltration and decreased myeloid-derived suppressor cell enrichment.Conclusions BANF1 is a key regulator of antitumor immunity mediated by cGAS-STING pathway. Therefore, our study provides a rational that targeting BANF1 is a potent strategy for enhancing immunotherapy for cancer with BANF1 upregulation.
first_indexed 2024-03-12T11:26:38Z
format Article
id doaj.art-cd515017455642a28a603fce88caff4a
institution Directory Open Access Journal
issn 2051-1426
language English
last_indexed 2024-03-12T11:26:38Z
publishDate 2023-08-01
publisher BMJ Publishing Group
record_format Article
series Journal for ImmunoTherapy of Cancer
spelling doaj.art-cd515017455642a28a603fce88caff4a2023-09-01T08:50:08ZengBMJ Publishing GroupJournal for ImmunoTherapy of Cancer2051-14262023-08-0111810.1136/jitc-2023-007035Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockadeFei Wang0Yang Li1Jinming Yu2Dawei Chen3Fei Wu4Meng Wu5Minglei Wang6Yiheng Huang7Minxin Chen8Weiyan Wang9Tao Zhong10Xiaozheng Chen11Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaSchool of Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaDepartment of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, ChinaBackground BANF1 is well known as a natural opponent of cyclic GMP-AMP synthase (cGAS) activity on genomic self-DNA. However, the roles of BANF1 in tumor immunity remain unclear. Here, we investigate the possible impact of BANF1 on antitumor immunity and response to immunotherapy.Methods The Cancer Genome Atlas public data were analyzed to evaluate the relevance of the expression of BANF1, patients’ survival and immune cell infiltration. We monitored tumor growth and explored the antitumor efficacy of targeting tumor-intrinsic BANF1 in combination with anti-programmed cell death protein-1 (PD-1) in MC38 or B16F10 tumor models in both immunocompetent and immunodeficient mice. Flow cytometry, immunofluorescence and T cells depletion experiments were used to validate the role of BANF1 in tumor immune microenvironment reprogramming. RNA sequencing was then used to interrogate the mechanisms how BANF1 regulated antitumor immunity.Results We show that upregulated expression of BANF1 in tumor tissues is significantly associated with poor survival and is negatively correlated with immune cell infiltration. Deficiency of BANF1 in tumor cells markedly antagonizes tumor growth in immunocompetent but not immunocompromised mice, and enhances the response to immunotherapy in murine models of melanoma and colon cancer. In the immunotherapy clinical cohort, patients with high BANF1 expression had a worse prognosis. Mechanistically, BANF1 knockout activates antitumor immune responses mediated by cGAS-synthase-stimulator of interferon genes (cGAS-STING) pathway, resulting in an immune-activating tumor microenvironment including increased CD8+ T cell infiltration and decreased myeloid-derived suppressor cell enrichment.Conclusions BANF1 is a key regulator of antitumor immunity mediated by cGAS-STING pathway. Therefore, our study provides a rational that targeting BANF1 is a potent strategy for enhancing immunotherapy for cancer with BANF1 upregulation.https://jitc.bmj.com/content/11/8/e007035.full
spellingShingle Fei Wang
Yang Li
Jinming Yu
Dawei Chen
Fei Wu
Meng Wu
Minglei Wang
Yiheng Huang
Minxin Chen
Weiyan Wang
Tao Zhong
Xiaozheng Chen
Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockade
Journal for ImmunoTherapy of Cancer
title Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockade
title_full Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockade
title_fullStr Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockade
title_full_unstemmed Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockade
title_short Inhibition of tumor intrinsic BANF1 activates antitumor immune responses via cGAS-STING and enhances the efficacy of PD-1 blockade
title_sort inhibition of tumor intrinsic banf1 activates antitumor immune responses via cgas sting and enhances the efficacy of pd 1 blockade
url https://jitc.bmj.com/content/11/8/e007035.full
work_keys_str_mv AT feiwang inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT yangli inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT jinmingyu inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT daweichen inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT feiwu inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT mengwu inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT mingleiwang inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT yihenghuang inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT minxinchen inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT weiyanwang inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT taozhong inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade
AT xiaozhengchen inhibitionoftumorintrinsicbanf1activatesantitumorimmuneresponsesviacgasstingandenhancestheefficacyofpd1blockade