细菌和癌细胞膜融合脂质体与 PD-L1 抑制剂配合用于癌症免疫疗法

IF 9.5 2区 材料科学 Q1 CHEMISTRY, PHYSICAL
Xianjin Luo, Chenglong Li, Zhaofei Guo, Hairui Wang, Penghui He, Yuanhao Zhao, Yi Lin, Chunting He, Yingying Hou, Yongshun Zhang, Guangsheng Du
{"title":"细菌和癌细胞膜融合脂质体与 PD-L1 抑制剂配合用于癌症免疫疗法","authors":"Xianjin Luo, Chenglong Li, Zhaofei Guo, Hairui Wang, Penghui He, Yuanhao Zhao, Yi Lin, Chunting He, Yingying Hou, Yongshun Zhang, Guangsheng Du","doi":"10.1007/s12274-024-6861-5","DOIUrl":null,"url":null,"abstract":"<p>Although tumor cell membranes with broad-spectrum antigens have been explored for cancer vaccines for decades, their relatively poor capacity to stimulate immune responses, especially cellular immune responses, has limited their application. Here, we presented a novel bacterial and cancerous cell membrane fusogenic liposome for co-delivering cell membrane-derived antigens and adjuvants. Meanwhile, a programmed death-ligand 1 (PD-L1) inhibitor, JQ-1, was incorporated into the formulation to tackle the up-regulated PD-L1 expression of antigen-presenting cells (APCs) upon vaccination, thereby augmenting its antitumor efficacy. The fusogenic liposomes demonstrated significantly improved cellular uptake by APCs and effectively suppressed PD-L1 expression in bone marrow-derived dendritic cells (BMDCs) <i>in vitro</i>. Following subcutaneous vaccination, the nanovaccines efficiently drained to the tumor-draining lymph nodes (TDLNs), and significantly inhibited PD-L1 expression of both dendritic cells (DCs) and macrophages within the TDLNs and tumors. As a result, the liposomal vaccine induced robust innate and cellular immune responses and inhibited tumor growth in a colorectal carcinoma-burden mouse model. In summary, the fabricated cell membrane-based fusogenic liposomes offer a safe, effective, and easily applicable strategy for tumor immunotherapy and hold potential for personalized cancer immunotherapy.\n</p>","PeriodicalId":713,"journal":{"name":"Nano Research","volume":null,"pages":null},"PeriodicalIF":9.5000,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Bacterial and cancerous cell membrane fused liposome coordinates with PD-L1 inhibitor for cancer immunotherapy\",\"authors\":\"Xianjin Luo, Chenglong Li, Zhaofei Guo, Hairui Wang, Penghui He, Yuanhao Zhao, Yi Lin, Chunting He, Yingying Hou, Yongshun Zhang, Guangsheng Du\",\"doi\":\"10.1007/s12274-024-6861-5\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Although tumor cell membranes with broad-spectrum antigens have been explored for cancer vaccines for decades, their relatively poor capacity to stimulate immune responses, especially cellular immune responses, has limited their application. Here, we presented a novel bacterial and cancerous cell membrane fusogenic liposome for co-delivering cell membrane-derived antigens and adjuvants. Meanwhile, a programmed death-ligand 1 (PD-L1) inhibitor, JQ-1, was incorporated into the formulation to tackle the up-regulated PD-L1 expression of antigen-presenting cells (APCs) upon vaccination, thereby augmenting its antitumor efficacy. The fusogenic liposomes demonstrated significantly improved cellular uptake by APCs and effectively suppressed PD-L1 expression in bone marrow-derived dendritic cells (BMDCs) <i>in vitro</i>. Following subcutaneous vaccination, the nanovaccines efficiently drained to the tumor-draining lymph nodes (TDLNs), and significantly inhibited PD-L1 expression of both dendritic cells (DCs) and macrophages within the TDLNs and tumors. As a result, the liposomal vaccine induced robust innate and cellular immune responses and inhibited tumor growth in a colorectal carcinoma-burden mouse model. In summary, the fabricated cell membrane-based fusogenic liposomes offer a safe, effective, and easily applicable strategy for tumor immunotherapy and hold potential for personalized cancer immunotherapy.\\n</p>\",\"PeriodicalId\":713,\"journal\":{\"name\":\"Nano Research\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":9.5000,\"publicationDate\":\"2024-08-03\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Nano Research\",\"FirstCategoryId\":\"88\",\"ListUrlMain\":\"https://doi.org/10.1007/s12274-024-6861-5\",\"RegionNum\":2,\"RegionCategory\":\"材料科学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CHEMISTRY, PHYSICAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Nano Research","FirstCategoryId":"88","ListUrlMain":"https://doi.org/10.1007/s12274-024-6861-5","RegionNum":2,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, PHYSICAL","Score":null,"Total":0}
引用次数: 0

摘要

尽管几十年来人们一直在探索将带有广谱抗原的肿瘤细胞膜用于癌症疫苗,但它们刺激免疫反应,尤其是细胞免疫反应的能力相对较差,这限制了它们的应用。在这里,我们提出了一种新型细菌和癌细胞膜融合脂质体,用于共同递送细胞膜衍生抗原和佐剂。同时,我们在配方中加入了程序性死亡配体1(PD-L1)抑制剂JQ-1,以解决接种疫苗后抗原递呈细胞(APCs)PD-L1表达上调的问题,从而增强其抗肿瘤功效。溶解脂质体明显提高了抗原递呈细胞(APCs)的细胞吸收率,并在体外有效抑制了骨髓树突状细胞(BMDCs)的 PD-L1 表达。皮下注射疫苗后,纳米疫苗有效地排泄到肿瘤引流淋巴结(TDLNs),并显著抑制了TDLNs和肿瘤内树突状细胞(DCs)和巨噬细胞的PD-L1表达。因此,脂质体疫苗诱导了强大的先天性免疫反应和细胞免疫反应,并抑制了结直肠癌负担小鼠模型中的肿瘤生长。总之,基于细胞膜的融合脂质体为肿瘤免疫疗法提供了一种安全、有效、易于应用的策略,并为个性化癌症免疫疗法提供了潜力。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Bacterial and cancerous cell membrane fused liposome coordinates with PD-L1 inhibitor for cancer immunotherapy

Bacterial and cancerous cell membrane fused liposome coordinates with PD-L1 inhibitor for cancer immunotherapy

Although tumor cell membranes with broad-spectrum antigens have been explored for cancer vaccines for decades, their relatively poor capacity to stimulate immune responses, especially cellular immune responses, has limited their application. Here, we presented a novel bacterial and cancerous cell membrane fusogenic liposome for co-delivering cell membrane-derived antigens and adjuvants. Meanwhile, a programmed death-ligand 1 (PD-L1) inhibitor, JQ-1, was incorporated into the formulation to tackle the up-regulated PD-L1 expression of antigen-presenting cells (APCs) upon vaccination, thereby augmenting its antitumor efficacy. The fusogenic liposomes demonstrated significantly improved cellular uptake by APCs and effectively suppressed PD-L1 expression in bone marrow-derived dendritic cells (BMDCs) in vitro. Following subcutaneous vaccination, the nanovaccines efficiently drained to the tumor-draining lymph nodes (TDLNs), and significantly inhibited PD-L1 expression of both dendritic cells (DCs) and macrophages within the TDLNs and tumors. As a result, the liposomal vaccine induced robust innate and cellular immune responses and inhibited tumor growth in a colorectal carcinoma-burden mouse model. In summary, the fabricated cell membrane-based fusogenic liposomes offer a safe, effective, and easily applicable strategy for tumor immunotherapy and hold potential for personalized cancer immunotherapy.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Nano Research
Nano Research 化学-材料科学:综合
CiteScore
14.30
自引率
11.10%
发文量
2574
审稿时长
1.7 months
期刊介绍: Nano Research is a peer-reviewed, international and interdisciplinary research journal that focuses on all aspects of nanoscience and nanotechnology. It solicits submissions in various topical areas, from basic aspects of nanoscale materials to practical applications. The journal publishes articles on synthesis, characterization, and manipulation of nanomaterials; nanoscale physics, electrical transport, and quantum physics; scanning probe microscopy and spectroscopy; nanofluidics; nanosensors; nanoelectronics and molecular electronics; nano-optics, nano-optoelectronics, and nano-photonics; nanomagnetics; nanobiotechnology and nanomedicine; and nanoscale modeling and simulations. Nano Research offers readers a combination of authoritative and comprehensive Reviews, original cutting-edge research in Communication and Full Paper formats. The journal also prioritizes rapid review to ensure prompt publication.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信