Tumor-derived autophagosome vaccine: mechanism of cross-presentation and therapeutic efficacy

Y Li, LX Wang, P Pang, Z Cui, S Aung, D Haley… - Clinical cancer …, 2011 - AACR
Y Li, LX Wang, P Pang, Z Cui, S Aung, D Haley, BA Fox, WJ Urba, HM Hu
Clinical cancer research, 2011AACR
Purpose: We previously reported that autophagy in tumor cells plays a critical role in cross-
presentation of tumor antigens and that autophagosomes are efficient antigen carriers for
cross-priming of tumor-reactive CD8+ T cells. Here, we sought to characterize further the
autophagosome-enriched vaccine named DRibble (DRiPs-containing blebs), which is
derived from tumor cells after inhibition of protein degradation, and to provide insights into
the mechanisms responsible for their efficacy as a novel cancer immunotherapy …
Abstract
Purpose: We previously reported that autophagy in tumor cells plays a critical role in cross-presentation of tumor antigens and that autophagosomes are efficient antigen carriers for cross-priming of tumor-reactive CD8+ T cells. Here, we sought to characterize further the autophagosome-enriched vaccine named DRibble (DRiPs-containing blebs), which is derived from tumor cells after inhibition of protein degradation, and to provide insights into the mechanisms responsible for their efficacy as a novel cancer immunotherapy.
Experimental Design: DRibbles were characterized by Western blot and light or transmission electron microscopy. The efficiency of cross-presentation mediated by DRibbles was first compared with that of whole-tumor cells and pure proteins. The mechanisms of antigen cross-presentation by DRibbles were analyzed, and the antitumor efficacy of the DRibble vaccine was tested in 3LL Lewis lung tumors and B16F10 melanoma.
Results: The DRibbles sequester both long-lived and short-lived proteins, including defective ribosomal products (DRiP), and damage-associated molecular pattern molecules exemplified by HSP90, HSP94, calreticulin, and HMGB1. DRibbles express ligands for CLEC9A, a newly described C-type lectin receptor expressed by a subset of conventional and plasmacytoid dendritic cells (DC), and cross-presentation was partially CLEC9A dependent. Furthermore, this autophagy-assisted antigen cross-presentation pathway involved both caveolae- and clathrin-mediated endocytosis and endoplasmic reticulum–associated degradation machinery. It depends on proteasome and TAP1, but not lysosome functions of antigen-presenting cells. Importantly, DCs loaded with autophagosome-enriched DRibbles can eradicate 3LL Lewis lung tumors and significantly delay the growth of B16F10 melanoma.
Conclusions: These data documented the unique characteristics and potent antitumor efficacy of the autophagosome-based DRibble vaccine. The efficacy of DRibble cancer vaccine will be further tested in clinical trials. Clin Cancer Res; 17(22); 7047–57. ©2011 AACR.
AACR