Preinduced reovirus-specific T-cell immunity enhances the anticancer efficacy of reovirus therapy

Christianne Groeneveldt, Priscilla Kinderman, Jordi J. C. van Stigt Thans, Camilla Labrie, Lisa Griffioen, Marjolein Sluijter, Diana J. M. van den Wollenberg, Rob C. Hoeben, Joke M. M. den Haan, Sjoerd H. van der Burg, Thorbald van Hall, Nadine van Montfoort

Research output: Contribution to journalArticleAcademicpeer-review

6 Citations (Scopus)

Abstract

BACKGROUND: Many solid tumors do not respond to immunotherapy due to their immunologically cold tumor microenvironment (TME). We and others found that oncolytic viruses (OVs), including reovirus type 3 Dearing, can enhance the efficacy of immunotherapy by recruiting CD8+ T cells to the TME. A significant part of the incoming CD8+ T cells is directed toward reovirus itself, which may be detrimental to the efficacy of OVs. However, here we aim to exploit these incoming virus-specific T cells as anticancer effector cells. METHODS: We performed an in-depth characterization of the reovirus-induced T-cell response in immune-competent mice bearing pancreatic KPC3 tumors. The immunodominant CD8+ T-cell epitope of reovirus was identified using epitope prediction algorithms and peptide arrays, and the quantity and quality of reovirus-specific T cells after reovirus administration were assessed using high-dimensional flow cytometry. A synthetic long peptide (SLP)-based vaccination strategy was designed to enhance the intratumoral frequency of reovirus-specific CD8+ T cells. RESULTS: Reovirus administration did not induce tumor-specific T cells but rather induced high frequencies of reovirus-specific CD8+ T cells directed to the immunodominant epitope. Priming of reovirus-specific T cells required a low-frequent population of cross-presenting dendritic cells which was absent in Batf3-/- mice. While intratumoral and intravenous reovirus administration induced equal systemic frequencies of reovirus-specific T cells, reovirus-specific T cells were highly enriched in the TME exclusively after intratumoral administration. Here, they displayed characteristics of potent effector cells with high expression of KLRG1, suggesting they may be responsive against local reovirus-infected cells. To exploit these reovirus-specific T cells as anticancer effector cells, we designed an SLP-based vaccination strategy to induce a strong T-cell response before virotherapy. These high frequencies of circulating reovirus-specific T cells were reactivated on intratumoral reovirus administration and significantly delayed tumor growth. CONCLUSIONS: These findings provide proof of concept that OV-specific T cells, despite not being tumor-specific, can be exploited as potent effector cells for anticancer treatment when primed before virotherapy. This is an attractive strategy for low-immunogenic tumors lacking tumor-specific T cells.
Original languageEnglish
Article numbere004464
JournalJournal for Immunotherapy of Cancer
Volume10
Issue number7
DOIs
Publication statusPublished - 19 Jul 2022

Keywords

  • Epitope Mapping
  • Immunotherapy
  • Lymphocytes, Tumor-Infiltrating
  • Oncolytic Viruses
  • Vaccination

Cite this