RNA aggregates harness the danger response for potent cancer immunotherapy

Hector R. Mendez-Gomez, Anna DeVries,Paul Castillo, Christina von Roemeling,Sadeem Qdaisat, Brian D. Stover, Chao Xie,Frances Weidert, Chong Zhao, Rachel Moor,Ruixuan Liu, Dhruvkumar Soni,Elizabeth Ogando-Rivas, Jonathan Chardon-Robles,James McGuiness,Dingpeng Zhang,Michael C. Chung, Christiano Marconi, Stephen Michel, Arnav Barpujari

Cell(2024)

引用 0|浏览2
暂无评分
摘要
Cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Here, we create “onion-like” multi-lamellar RNA lipid particle aggregates (LPAs) to substantially enhance the payload packaging and immunogenicity of tumor mRNA antigens. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for Toll-like receptor engagement in immune cells, systemically administered RNA-LPAs activate RIG-I in stromal cells, eliciting massive cytokine/chemokine response and dendritic cell/lymphocyte trafficking that provokes cancer immunogenicity and mediates rejection of both early- and late-stage murine tumor models. In client-owned canines with terminal gliomas, RNA-LPAs improved survivorship and reprogrammed the TME, which became “hot” within days of a single infusion. In a first-in-human trial, RNA-LPAs elicited rapid cytokine/chemokine release, immune activation/trafficking, tissue-confirmed pseudoprogression, and glioma-specific immune responses in glioblastoma patients. These data support RNA-LPAs as a new technology that simultaneously reprograms the TME while eliciting rapid and enduring cancer immunotherapy.
更多
查看译文
关键词
mRNA,cancer vaccines,lipid nanoparticles,cancer immunotherapy,glioblastoma,personalized therapy,translational therapeutics,brain tumors
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要