Epigenetic modulation enhances immunotherapy for hepatocellular carcinoma

•PD1-inhibitor nivolumab has marginal objective response rate of 20% in HCC.•Epigenetic therapy can potentiate immunotherapy for HCC.•Strategy: Target EZH2 and DNMT1 with anti-PDL-1 monoclonal antibodies.•Mechanisms: increased neoantigen expression, cytotoxic T-cell infiltration, and apoptosis. Anti...

Full description

Saved in:
Bibliographic Details
Published inCellular immunology Vol. 336; pp. 66 - 74
Main Authors Hong, Young K., Li, Yan, Pandit, Harshul, Li, SuPing, Pulliam, Zachary, Zheng, Qianqian, Yu, Youxi, Martin, Robert C.G.
Format Journal Article
LanguageEnglish
Published Netherlands Elsevier Inc 01.02.2019
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:•PD1-inhibitor nivolumab has marginal objective response rate of 20% in HCC.•Epigenetic therapy can potentiate immunotherapy for HCC.•Strategy: Target EZH2 and DNMT1 with anti-PDL-1 monoclonal antibodies.•Mechanisms: increased neoantigen expression, cytotoxic T-cell infiltration, and apoptosis. Anti-PDL-1 immunotherapy for Hepatocellular Carcinoma (HCC) demonstrated a mixed response. Polycomb Repressor Complex 2(PRC2) contributes to the initiation and progression of HCC by suppressing tumor antigens and inhibiting an immune response. Two components of epigenetic modulation are Enhancer of Zeste Homolog 2 (EZH2, the catalytic component of PRC2) and DNA Methyltransferase 1 (DNMT1). We aim to investigate the potential role of epigenetic therapy targeting EZH2 and DNMT1 as a novel strategy to modulate immunotherapy response in HCC. HepG2, Hep3B, and Hepa1-6 HCC cell lines were treated with EZH2 inhibitor (DZNep) and DNMT1 inhibitor (5-Azacytidine) with and without anti-PDL-1. Quantitative RT-PCR and immunohistochemistry were performed to evaluate the expression of tumor suppressors, tumor antigens, and Th1 chemokines. In-vivo C57/LJ immunocompetent mice model with subcutaneous tumor inoculation was performed with intraperitoneal drug injections. There was a significant upregulation of Th1 chemokines in HepG2 (CXCL9 5.5 ± 0.2 relative fold change; CXCL10 1.44 × 103 ± 37 relative fold change) and Hep3B (CXCL 9 6.85 × 103 ± 1.3 × 103 relative fold change; CXCL 10 2.15 × 103 ± 3.1 × 102 relative fold change). Additionally, there was a significant induction of cancer testis antigens NY-ESO-1 (3.6–3.7 ± 0.3 relative fold change) and LAGE (8.3–11.7 ± 1.9 relative fold change). In vivo model demonstrated statistically significant tumor regression in the combination treatment group (0.02 g ± 0.02) compared to epigenetic therapy (0.63 g ± 0.61) or immunotherapy alone (0.15 g ± 0.21) with untreated control (2.4 g ± 0.71). There was significantly increased trafficking of cytotoxic T- lymphocytes and associated apoptosis for the combination treatment group compared to epigenetic or immunotherapy alone. This study demonstrates that epigenetic modulation could be a novel potential strategy to augment immunotherapy for HCC by stimulating T cell trafficking into tumor microenvironment via activation of transcriptionally repressed chemokine genes responsible for T-cell trafficking, inducing previously silent neoantigens for immune targets, and allowing tumor regression as a result. A clinical trial of this feasible combination therapy of these clinically available agents is warranted.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:0008-8749
1090-2163
DOI:10.1016/j.cellimm.2018.12.010