1. Academic Validation
  2. Low dose decitabine treatment induces CD80 expression in cancer cells and stimulates tumor specific cytotoxic T lymphocyte responses

Low dose decitabine treatment induces CD80 expression in cancer cells and stimulates tumor specific cytotoxic T lymphocyte responses

  • PLoS One. 2013 May 9;8(5):e62924. doi: 10.1371/journal.pone.0062924.
Li-Xin Wang 1 Zhen-Yang Mei Ji-Hao Zhou Yu-Shi Yao Yong-Hui Li Yi-Han Xu Jing-Xin Li Xiao-Ning Gao Min-Hang Zhou Meng-Meng Jiang Li Gao Yi Ding Xue-Chun Lu Jin-Long Shi Xu-Feng Luo Jia Wang Li-Li Wang Chunfeng Qu Xue-Feng Bai Li Yu
Affiliations

Affiliation

  • 1 Department of Hematology, Chinese PLA General Hospital, Beijing, China.
Abstract

Lack of immunogenicity of Cancer cells has been considered a major reason for their failure in induction of a tumor specific T cell response. In this paper, we present evidence that decitabine (DAC), a DNA methylation inhibitor that is currently used for the treatment of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and other malignant neoplasms, is capable of eliciting an anti-tumor cytotoxic T lymphocyte (CTL) response in mouse EL4 tumor model. C57BL/6 mice with established EL4 tumors were treated with DAC (1.0 mg/kg body weight) once daily for 5 days. We found that DAC treatment resulted in infiltration of IFN-γ producing T lymphocytes into tumors and caused tumor rejection. Depletion of CD8(+), but not CD4(+) T cells resumed tumor growth. DAC-induced CTL response appeared to be elicited by the induction of CD80 expression on tumor cells. Epigenetic evidence suggests that DAC induces CD80 expression in EL4 cells via demethylation of CpG dinucleotide sites in the promoter of CD80 gene. In addition, we also showed that a transient, low-dose DAC treatment can induce CD80 gene expression in a variety of human Cancer cells. This study provides the first evidence that epigenetic modulation can induce the expression of a major T cell co-stimulatory molecule on Cancer cells, which can overcome immune tolerance, and induce an efficient anti-tumor CTL response. The results have important implications in designing DAC-based Cancer Immunotherapy.

Figures
Products