Your browser doesn't support javascript.
loading
Histone Deacetylase Inhibitors Directly Modulate T Cell Gene Expression and Signaling and Promote Development of Effector-Exhausted T Cells in Murine Tumors.
Ibrahim, Mohammed L; Zheng, Hong; Barlow, Margaret L; Latif, Yousuf; Chen, Zhihua; Yu, Xiaoqing; Beg, Amer A.
Afiliación
  • Ibrahim ML; Department of Immunology, Moffitt Cancer Center, Tampa, FL.
  • Zheng H; Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
  • Barlow ML; Department of Immunology, Moffitt Cancer Center, Tampa, FL.
  • Latif Y; Department of Immunology, Moffitt Cancer Center, Tampa, FL.
  • Chen Z; Department of Immunology, Moffitt Cancer Center, Tampa, FL.
  • Yu X; Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL.
  • Beg AA; Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL.
J Immunol ; 212(4): 737-747, 2024 Feb 15.
Article en En | MEDLINE | ID: mdl-38169329
ABSTRACT
Epigenetic regulation plays a crucial role in the development and progression of cancer, including the regulation of antitumor immunity. The reversible nature of epigenetic modifications offers potential therapeutic avenues for cancer treatment. In particular, histone deacetylase (HDAC) inhibitors (HDACis) have been shown to promote antitumor T cell immunity by regulating myeloid cell types, enhancing tumor Ag presentation, and increasing expression of chemokines. HDACis are currently being evaluated to determine whether they can increase the response rate of immune checkpoint inhibitors in cancer patients. Although the potential direct effect of HDACis on T cells likely impacts antitumor immunity, little is known about how HDAC inhibition alters the transcriptomic profile of T cells. In this article, we show that two clinical-stage HDACis profoundly impact gene expression and signaling networks in CD8+ and CD4+ T cells. Specifically, HDACis promoted T cell effector function by enhancing expression of TNF-α and IFN-γ and increasing CD8+ T cell cytotoxicity. Consistently, in a murine tumor model, HDACis led to enrichment of CD8+ T cell subsets with high expression of effector molecules (Prf1, Ifng, Gzmk, and Grmb) but also molecules associated with T cell exhaustion (Tox, Pdcd1, Lag3, and Havcr2). HDACis further generated a tumor microenvironment dominated by myeloid cells with immune suppressive signatures. These results indicate that HDACis directly and favorably augment T cell effector function but also increase their exhaustion signal in the tumor microenvironment, which may add a layer of complexity for achieving clinical benefit in combination with immune checkpoint inhibitors.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Inhibidores de Histona Desacetilasas / Neoplasias Límite: Animals / Humans Idioma: En Revista: J Immunol Año: 2024 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Inhibidores de Histona Desacetilasas / Neoplasias Límite: Animals / Humans Idioma: En Revista: J Immunol Año: 2024 Tipo del documento: Article