Abstract

Triple-negative breast cancer (TNBC) represents a more aggressive and difficult subtype of breast cancer where responses to chemotherapy occur, but toxicity is significant and resistance often follows. Immunotherapy has shown promising results in various types of cancer, including breast cancer. Here, we investigated a new combination strategy where histone deacetylase inhibitors (HDACi) are applied with immune checkpoint inhibitors to improve immunotherapy responses in TNBC.Testing different epigenetic modifiers, we focused on the mechanisms underlying HDACi as priming modulators of immunotherapy. Tumor cells were co-cultured with human peripheral blood mononuclear cells (PBMCs) and flow cytometric immunophenotyping was performed to define the role of epigenetic priming in promoting tumor antigen presentation and immune cell activation. We found that HDACi up-regulate PD-L1 mRNA and protein expression in a time-dependent manner in TNBC cells, but not in hormone responsive cells. Focusing on TNBC, HDACi up-regulated PD-L1 and HLA-DR on tumor cells when co-cultured with PBMCs and down-regulated CD4+ Foxp3+ Treg in vitro. HDACi significantly enhanced the in vivo response to PD-1/CTLA-4 blockade in the triple-negative 4T1 breast cancer mouse model, the only currently available experimental system with functional resemblance to human TNBC. This resulted in a significant decrease in tumor growth and increased survival, associated with increased T cell tumor infiltration and a reduction in CD4+ Foxp3+ T cells in the tumor microenvironment. Overall, our results suggest a novel role for HDAC inhibition in combination with immune checkpoint inhibitors and identify a promising therapeutic strategy, supporting its further clinical evaluation for TNBC treatment.

Highlights

  • Breast cancer is one of the most common diseases, second only to lung cancer as the leading cause of cancer death in women, accounting for 30% of new diagnoses [1]

  • These effects were observed with class non-specific Histone deacetylase inhibitors (HDACi), and with class specific HDACi such as valproic acid (VPA) and entinostat (Figure 1B)

  • Previous reports have shown that HDACi modulate immune response, alter regulatory T cells (Treg) activity and regulate cytokine expression [6, 7, 37, 38]

Read more

Summary

Introduction

Breast cancer is one of the most common diseases, second only to lung cancer as the leading cause of cancer death in women, accounting for 30% of new diagnoses [1]. Histone deacetylase inhibitors (HDACi) represent a new class of anticancer agents that induce a wide range of transient gene expression alterations, without www.impactjournals.com/oncotarget implicating permanent changes in DNA sequence [4, 5]. The cellular response to HDACi is complex and involves the regulation of histone and non-histone proteins by modifying their post-translational acetylation, playing a critical role in various cancer pathways. In addition to their effects on cancer signaling, HDACi have distinct immune modulatory functions, including modulation of regulatory T cells (Treg), Foxp expression and changes in tumor-infiltrating lymphocytes (TILs) composition [6, 7]. Emerging data suggest that epigenetic modulation is important for controlling T cells infiltration, differentiation, and PD-L1 expression [7,8,9]

Methods
Results
Conclusion
Full Text
Published version (Free)

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