Abstract

Immune checkpoint blockade (ICB), particularly programmed death 1 (PD-1) and its ligand (PD-L1), has shown considerable clinical benefits in patients with various cancers. Many studies show that PD-L1 expression may be biomarkers to help select responders for anti-PD-1 treatment. Therefore, it is necessary to elucidate the molecular mechanisms that control PD-L1 expression. As a potential chemosensitizer and anticancer drug, disulfiram (DSF) kills tumor cells via regulating multiple signaling pathways and transcription factors. However, its effect on tumor immune microenvironment (TIME) remains unclear. Here, we showed that DSF increased PD-L1 expression in triple negative breast cancer (TNBC) cells. Through bioinformatics analysis, we found that DNMT1 was highly expressed in TNBC tissue and PD-L1 was negatively correlated with IRF7 expression. DSF reduced DNMT1 expression and activity, and hypomethylated IRF7 promoter region resulting in upregulation of IRF7. Furthermore, we found DSF enhanced PD-L1 expression via DNMT1-mediated IRF7 hypomethylation. In in vivo experiments, DSF significantly improved the response to anti-PD-1 antibody (Ab) in 4T1 breast cancer mouse model. Immunohistochemistry staining showed that granzyme B+ and CD8+ T cells in the tumor tissues were significantly increased in the combination group. By analyzing the results of the tumor tissue RNA sequencing, four immune-associated pathways were significantly enriched in the DSF joint anti-PD-1 Ab group. In conclusion, we found that DSF could upregulate PD-L1 in TNBC cells and elucidated its mechanism. Our findings revealed that the combination of DSF and anti-PD-1 Ab could activate TIME to show much better antitumor efficacy than monotherapy.

Highlights

  • Breast cancer (BC) is one of the most frequent diseases and the leading cause of cancer death among females [1]

  • Despite anti-programmed death 1 (PD-1) therapy has shown promising clinical benefits in patients with triple negative BC (TNBC), a significant fraction of patients remains unresponsive to this therapy

  • interferon regulatory factor 7 (IRF7) is an important regulator of type I IFN responses, and its transactivation can amplify the generation of type I IFNs via a positive feedback loop [43]

Read more

Summary

Introduction

Breast cancer (BC) is one of the most frequent diseases and the leading cause of cancer death among females [1]. There is an urgent need to explore novel therapeutic strategies to improve the clinical outcomes of TNBC patients. In the tumor microenvironment (TME), inducing PD-L1 expression can lead to PD-1-mediated T cell exhaustion, suppressing the antitumor cytotoxic T cell response [7]. Such negative interaction can be inhibited by anti-PD-1/anti-PD-L1 antibodies (Abs). PD-1/PD-L1 checkpoint blockades have been approved by the FDA in various cancers, including lung cancer, Hodgkin lymphoma, and BC [8,9,10]. Improving the therapeutic effect of anti-PD-1 antibody (Ab) in TNBC patients is urgent and valuable

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.