新抗原mRNA疫苗可诱导祖细胞耗竭的T细胞,这些细胞可支持对伴有腹膜转移的胃癌进行抗PD-1治疗。

Neoantigen mRNA vaccines induce progenitor-exhausted T cells that support anti-PD-1 therapy in gastric cancer with peritoneal metastasis.

作者信息

Nagaoka Koji, Nakanishi Hideyuki, Tanaka Hiroki, Anindita Jessica, Kawamura Takeshi, Tanaka Toshiya, Yamashita Takefumi, Kuroda Akihiro, Nomura Sachiyo, Akita Hidetaka, Itaka Keiji, Kodama Tatsuhiko, Kakimi Kazuhiro

机构信息

Department of Immunology, Kindai University, Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-0014, Japan.

Department of Biofunction Research, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research, Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-Ku, Tokyo, 101-0062, Japan.

出版信息

Gastric Cancer. 2025 Jul 31. doi: 10.1007/s10120-025-01640-8.

Abstract

BACKGROUND

Gastric cancer with peritoneal metastasis is associated with a poor prognosis. Current treatments, including the first-line therapy of combination chemotherapy with nivolumab for advanced recurrent gastric cancer, have shown limited efficacy against peritoneal dissemination. In this study, we evaluated neoantigen (neoAg)-mRNA lipid nanoparticle (LNP) as a potential agent in combination with anti-PD-1 therapy, focusing on its effects on neoAg-specific CD8 T cell responses and antitumor efficacy in a murine gastric cancer model.

METHODS

The mRNA, comprising a tandem minigene encoding three neoAgs identified from the murine gastric cancer YTN16 cell line, was synthesized by in vitro transcription and encapsulated within LNPs. NeoAg-specific CD8 T cells in the spleens and tumors were assessed by flow cytometry. The antitumor efficacy of the neoAg-mRNA-LNP vaccine, alone or in combination with anti-PD-1 antibody, was evaluated in both subcutaneous and peritoneal metastasis models of YTN16.

RESULTS

The neoAg-mRNA-LNP vaccine induced significantly higher frequencies of neoAg-specific CD8 T cells than the neoAg-dendritic cell vaccine, confirming its enhanced immunogenicity. NeoAg-mRNA-LNP vaccination led to robust tumor regression, achieving complete eradication in all treated mice, especially when combined with anti-PD-1 therapy. This effect was associated with an increase in neoAg-specific progenitor-exhausted and intermediate-exhausted CD8 T cells. In a peritoneal metastasis model, neoAg-mRNA-LNP monotherapy prevented peritoneal dissemination when administered prophylactically, and combination therapy with anti-PD-1 effectively suppressed tumor growth in a therapeutic setting.

CONCLUSIONS

NeoAg-mRNA-LNP vaccines elicit potent neoAg-specific CD8 T cell responses and show enhanced antitumor efficacy with anti-PD-1 therapy in gastric cancer with peritoneal metastasis.

摘要

背景

伴有腹膜转移的胃癌预后较差。目前的治疗方法,包括晚期复发性胃癌一线使用纳武单抗联合化疗,对腹膜播散的疗效有限。在本研究中,我们评估了新抗原(neoAg)-信使核糖核酸(mRNA)脂质纳米颗粒(LNP)作为一种与抗程序性死亡蛋白1(PD-1)治疗联合使用的潜在药物,重点关注其对新抗原特异性CD8 T细胞反应的影响以及在小鼠胃癌模型中的抗肿瘤疗效。

方法

通过体外转录合成包含串联小基因的mRNA,该串联小基因编码从鼠胃癌YTN16细胞系中鉴定出的三种新抗原,并将其封装在LNP中。通过流式细胞术评估脾脏和肿瘤中新抗原特异性CD8 T细胞。在YTN16的皮下和腹膜转移模型中评估新抗原-mRNA-LNP疫苗单独或与抗PD-1抗体联合使用的抗肿瘤疗效。

结果

与新抗原-树突状细胞疫苗相比,新抗原-mRNA-LNP疫苗诱导的新抗原特异性CD8 T细胞频率显著更高,证实了其增强的免疫原性。新抗原-mRNA-LNP疫苗接种导致显著的肿瘤消退,在所有治疗小鼠中实现了完全根除,尤其是与抗PD-1治疗联合使用时。这种效应与新抗原特异性祖细胞耗竭和中度耗竭的CD8 T细胞增加有关。在腹膜转移模型中,新抗原-mRNA-LNP单药预防性给药可防止腹膜播散,在治疗环境中,与抗PD-1联合治疗可有效抑制肿瘤生长。

结论

新抗原-mRNA-LNP疫苗在伴有腹膜转移的胃癌中引发强大的新抗原特异性CD8 T细胞反应,并与抗PD-1治疗联合显示出增强的抗肿瘤疗效。

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索