• Medientyp: E-Artikel
  • Titel: A modular self-adjuvanting cancer vaccine combined with an oncolytic vaccine induces potent antitumor immunity
  • Beteiligte: Das, Krishna; Belnoue, Elodie; Rossi, Matteo; Hofer, Tamara; Danklmaier, Sarah; Nolden, Tobias; Schreiber, Liesa-Marie; Angerer, Katharina; Kimpel, Janine; Hoegler, Sandra; Spiesschaert, Bart; Kenner, Lukas; von Laer, Dorothee; Elbers, Knut; Derouazi, Madiha; Wollmann, Guido
  • Erschienen: Springer Science and Business Media LLC, 2021
  • Erschienen in: Nature Communications, 12 (2021) 1
  • Sprache: Englisch
  • DOI: 10.1038/s41467-021-25506-6
  • ISSN: 2041-1723
  • Entstehung:
  • Anmerkungen:
  • Beschreibung: <jats:title>Abstract</jats:title><jats:p>Functional tumor-specific cytotoxic T cells elicited by therapeutic cancer vaccination in combination with oncolytic viruses offer opportunities to address resistance to checkpoint blockade therapy. Two cancer vaccines, the self-adjuvanting protein vaccine KISIMA, and the recombinant oncolytic vesicular stomatitis virus pseudotyped with LCMV-GP expressing tumor-associated antigens, termed VSV-GP-TAA, both show promise as a single agent. Here we find that, when given in a heterologous prime-boost regimen with an optimized schedule and route of administration, combining KISIMA and VSV-GP-TAA vaccinations induces better cancer immunity than individually. Using several mouse tumor models with varying degrees of susceptibility for viral replication, we find that priming with KISIMA-TAA followed by VSV-GP-TAA boost causes profound changes in the tumor microenvironment, and induces a large pool of poly-functional and persistent antigen-specific cytotoxic T cells in the periphery. Combining this heterologous vaccination with checkpoint blockade further improves therapeutic efficacy with long-term survival in the spectrum. Overall, heterologous vaccination with KISIMA and VSV-GP-TAA could sensitize non-inflamed tumors to checkpoint blockade therapy.</jats:p>
  • Zugangsstatus: Freier Zugang