Chimeric Exosomes Functionalized with STING Activation for Personalized Glioblastoma Immunotherapy

ADVANCED SCIENCE(2024)

Cited 0|Views34
No score
Abstract
A critical challenge of existing cancer vaccines is to orchestrate the demands of antigen-enriched furnishment and optimal antigen-presentation functionality within antigen-presenting cells (APCs). Here, a complementary immunotherapeutic strategy is developed using dendritic cell (DC)-tumor hybrid cell-derived chimeric exosomes loaded with stimulator of interferon genes (STING) agonists (DT-Exo-STING) for maximized tumor-specific T-cell immunity. These chimeric carriers are furnished with broad-spectrum antigen complexes to elicit a robust T-cell-mediated inflammatory program through direct self-presentation and indirect DC-to-T immunostimulatory pathway. This chimeric exosome-assisted delivery strategy possesses the merits versus off-the-shelf cyclic dinucleotide (CDN) delivery techniques in both the brilliant tissue-homing capacity, even across the intractable blood-brain barrier (BBB), and the desired cytosolic entry for enhanced STING-activating signaling. The improved antigen-presentation performance with this nanovaccine-driven STING activation further enhances tumor-specific T-cell immunoresponse. Thus, DT-Exo-STING reverses immunosuppressive glioblastoma microenvironments to pro-inflammatory, tumoricidal states, leading to an almost obliteration of intracranial primary lesions. Significantly, an upscaling option that harnesses autologous tumor tissues for personalized DT-Exo-STING vaccines increases sensitivity to immune checkpoint blockade (ICB) therapy and exerts systemic immune memory against post-operative glioma recrudesce. These findings represent an emerging method for glioblastoma immunotherapy, warranting further exploratory development in the clinical realm. A complementary immunotherapeutic platform is constructed using DC-tumor chimeric exosomes cooperated with exogenous CDN-driven STING activation for maximized tumor-specific T-cell immunity. These chimeric nanovesicles provide advantages versus existing CDN delivery technologies in 1) the supply of broad-spectrum tumor antigen complexes for dual T-cell activation; 2) the cytosolic delivery of STING agonists for enhanced antigen presentation; and 3) the brilliant BBB-crossing capacity.image
More
Translated text
Key words
chimeric exosomes,glioblastoma,personalized immunotherapy,STING activation,T cell responses
AI Read Science
Must-Reading Tree
Example
Generate MRT to find the research sequence of this paper
Chat Paper
Summary is being generated by the instructions you defined