Abstract

Simple SummaryRadiotherapy (RT) with higher single doses is capable of creating a beneficial immunological tumor microenvironment for additional immune therapies to trigger systemic anti-tumor immune responses. Here, we aimed to boost RT-induced immune responses with inactivated whole tumor cell vaccines generated with high hydrostatic pressure (HHP) and to break immunosuppression by inhibition of suppressive immune checkpoint molecules, such as PD-1. Our results highlight that HHP vaccines act systemically to retard tumor growth, but only of previously irradiated tumors. In contrast, abscopal anti-tumor immune responses, namely those to non-irradiated tumors, are triggered by a combination of RT of not too many fractions plus anti-PD-1, being characterized by the induction of distinct immune alterations in the primary and abscopal tumor, respectively. This emphasizes that future multimodal radioimmunotherapy approaches need to be carefully optimized regarding RT fractionation and the interaction of different immunotherapies with each other.Radiotherapy (RT) is known to have immune-modulatory properties. We hypothesized that RT and inactivated whole tumor cell vaccines generated with high hydrostatic pressure (HHP) synergize to retard the tumor growth which can be additionally improved with anti-PD-1 treatment. In abscopal tumor models, we injected mice with B16-F10 melanoma or TS/A mammary tumors. To evaluate the efficiency of RT in combination with HHP vaccines, we locally irradiated only one tumor with 2 × 8 Gy or 3 × 8 Gy. HHP vaccines further retarded the growth of locally irradiated (2 × 8 Gy) tumors. However, HHP vaccination combined with RT failed to induce abscopal anti-tumor immune responses, namely those to non-irradiated tumors, and even partly abrogated those which were induced with RT plus anti-PD-1. In the latter group, the abscopal effects were accompanied by an elevated infiltration of CD8+ T cells, monocytes/macrophages, and dendritic cells. 3 × 8 Gy failed to induce abscopal effects in association with increased expression of immunosuppressive checkpoint molecules compared to 2 × 8 Gy. We conclude that HHP vaccines induce anti-tumor effects, but only if the tumor microenvironment was previously modulated by hypofractionated RT with not too many fractions, but failed to improve RT plus anti-PD-induced abscopal responses that are characterized by distinct immune alterations.

Highlights

  • The treatment of a tumor with ionizing radiation is known to induce immunogenic cell death in tumor cells under certain conditions, which is characterized by the release of damage-associated molecular patterns (DAMPs), tumor antigens, pro-inflammatory cytokines, and others, which turns them into an in situ cancer vaccine

  • We found that high hydrostatic pressure (HHP) vaccines work systemically, but only on previously irradiated tumors and that abscopal responses to RT plus anti-PD1 are even weakened by HHP

  • Based on the synergistic effect of RT and HHP vaccination we observed when injecting the vaccine peritumorally [21], we aimed to investigate if HHP vaccines can induce systemic immune responses after injection distant to the tumor

Read more

Summary

Introduction

The treatment of a tumor with ionizing radiation is known to induce immunogenic cell death in tumor cells under certain conditions, which is characterized by the release of damage-associated molecular patterns (DAMPs), tumor antigens, pro-inflammatory cytokines, and others, which turns them into an in situ cancer vaccine. In times of combination of RT with immunotherapies (IT), abscopal effects are related to anti-tumor effects of RT that are observed outside of the radiation field. They are mediated by the immune system [5]. ITs have great potential to complement RT by further boosting RT-induced immune responses and/or counteracting tumor-intrinsic as well as RT-mediated immunosuppression [9] This beneficial liaison of combinations of RT and ITs is reflected by the continuously increasing reports of abscopal effects since the approval of the immune checkpoint inhibitor ipilimumab in 2011 [10] and ITs in general [11]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call