Peptidic microarchitecture-trapped tumor vaccine combined with immune checkpoint inhibitor or PI3K gamma inhibitor can enhance immunogenicity and eradicate tumors
Du, Yang2,4; Liu, Ye1; Wang, Di3; Bai, Hua3; Wang, Zhijie3; He, Xiran2,3; Zhang, Pei2,3; Tian, Jie2,4,5,6; Wang, Jie3
刊名JOURNAL FOR IMMUNOTHERAPY OF CANCER
2022-02-01
卷号10期号:2页码:13
关键词immunotherapy immunogenicity vaccine tumor microenvironment lymphocytes tumor infiltrating macrophages
DOI10.1136/jitc-2021-003564
通讯作者Tian, Jie(jie.tian@ia.ac.cn) ; Wang, Jie(zlhuxi@163.com)
英文摘要Background With the rapid development of immune checkpoint inhibitors and neoantigen (NeoV)-based personalized tumor vaccines, tumor immunotherapy has shown promising therapeutic results. However, the limited efficacy of available tumor vaccines impedes the development of personalized tumor immunotherapy. In this study, we developed a novel tumor vaccine system and proposed combined therapeutic strategies for improving treatment effects. Methods We developed a novel tumor vaccine system comprising a newly synthesized peptidic microarchitecture (PMA) with high assembly efficacy. The PMA-trapped neoantigen vaccine was developed to codeliver tumor neoantigen and the Toll-like receptor 9 agonist CpG (NeoV), abbreviated as PMA-NeoV. A microfluidic chip was used to produce PMA particles in a uniform and precise manner. Vaccine effectiveness was investigated both in vitro and in vivo. The combined immunotherapeutic effect of PMA-NeoV with anti-programmed cell death ligand 1 antibody (aPD-L1) or with the phosphatidylinositol 3-kinase gamma (PI3K gamma) inhibitor IPI-549 was further tested in MC38 mouse tumor model. Results PMA-NeoV not only promoted codelivery of the tumor vaccine but also potentiated vaccine immunogenicity. Moreover, compared with free NeoV, PMA-NeoV significantly increased the number of tumor-infiltrating lymphocytes, promoted the neoantigen-specific systemic immune response, and suppressed murine colon MC38 tumor growth. Furthermore, PMA-NeoV increased the expression of programmed cell death receptor-1 on T lymphocytes, and in combination with aPD-L1 eradicated seven of eight MC38 tumors by rescuing exhausted T lymphocytes. Moreover, we combined the PMA-NeoV with the IPI-549, a molecular switch that controls immune suppression, and found that this combination significantly suppressed tumor growth and eradicated five of eight inoculated tumors, by switching suppressive macrophages to their active state and activating T cells to prime a robust tumor immune microenvironment. Conclusions We developed a tumor vaccine delivery system and presented a promising personalized tumor vaccine-based therapeutic regimen in which a tumor vaccine delivery system is combined with an aPD-L1 or PI3K gamma inhibitor to improve tumor immunotherapy outcomes.
资助项目Beijing Natural Science Foundation[7212207] ; National Natural Science Foundation of China[81871514] ; National Natural Science Foundation of China[92159303] ; National Natural Science Foundation of China[62027901] ; National Natural Science Foundation of China[81227901] ; National Natural Science Foundation of China[81470083] ; National Natural Science Foundation of China[81527805] ; Ministry of Science and Technology of China[2017YFA0205200] ; Ministry of Science and Technology of China[2017YFA0700401] ; National Key Research and Development Program of China[2017YFA0700401] ; Leading Medical Talents Program of Health Commission of Yunnan Province[L-2018013] ; Young and Middle Aged Academic and Technical Leaders Program of Yunnan Province[202005AC160010]
WOS关键词CANCER ; BLOCKADE ; RESPONSES ; MUTANOME ; EFFICACY ; CELLS
WOS研究方向Oncology ; Immunology
语种英语
出版者BMJ PUBLISHING GROUP
WOS记录号WOS:000762406500006
资助机构Beijing Natural Science Foundation ; National Natural Science Foundation of China ; Ministry of Science and Technology of China ; National Key Research and Development Program of China ; Leading Medical Talents Program of Health Commission of Yunnan Province ; Young and Middle Aged Academic and Technical Leaders Program of Yunnan Province
内容类型期刊论文
源URL[http://ir.ia.ac.cn/handle/173211/48032]  
专题自动化研究所_中国科学院分子影像重点实验室
通讯作者Tian, Jie; Wang, Jie
作者单位1.Chinese Acad Med Sci, Peking Union Med Coll, Inst Med Biol, Kunming, Yunnan, Peoples R China
2.Chinese Acad Sci, Inst Automat, CAS Key Lab Mol Imaging, Beijing Key Lab Mol Imaging,State Key Lab Managem, Beijing, Peoples R China
3.Chinese Acad Med Sci & Peking Union Med Coll, Natl Canc Ctr, Dept Med Oncol, Natl Clin Res Ctr Canc,Canc Hosp, Beijing, Peoples R China
4.Univ Chinese Acad Sci, Beijing, Peoples R China
5.Beihang Univ, Beijing Adv Innovat Ctr Big Data Based Precis Med, Sch Med, Beijing, Peoples R China
6.Xidian Univ, Sch Life Sci & Technol, Xian, Shaanxi, Peoples R China
推荐引用方式
GB/T 7714
Du, Yang,Liu, Ye,Wang, Di,et al. Peptidic microarchitecture-trapped tumor vaccine combined with immune checkpoint inhibitor or PI3K gamma inhibitor can enhance immunogenicity and eradicate tumors[J]. JOURNAL FOR IMMUNOTHERAPY OF CANCER,2022,10(2):13.
APA Du, Yang.,Liu, Ye.,Wang, Di.,Bai, Hua.,Wang, Zhijie.,...&Wang, Jie.(2022).Peptidic microarchitecture-trapped tumor vaccine combined with immune checkpoint inhibitor or PI3K gamma inhibitor can enhance immunogenicity and eradicate tumors.JOURNAL FOR IMMUNOTHERAPY OF CANCER,10(2),13.
MLA Du, Yang,et al."Peptidic microarchitecture-trapped tumor vaccine combined with immune checkpoint inhibitor or PI3K gamma inhibitor can enhance immunogenicity and eradicate tumors".JOURNAL FOR IMMUNOTHERAPY OF CANCER 10.2(2022):13.
个性服务
查看访问统计
相关权益政策
暂无数据
收藏/分享
所有评论 (0)
暂无评论
 

除非特别说明,本系统中所有内容都受版权保护,并保留所有权利。


©版权所有 ©2017 CSpace - Powered by CSpace