Abstract

BackgroundImmunogenic radiotherapy (RT) can act synergistically with immune checkpoint blockers (ICBs). However, alternatives are needed for non-responding patients and those with pre-existing or ICB-induced autoimmune symptoms. Combination of RT with IL-2 could be an alternative. But IL-2 has a short half-life, and, by binding to its high-affinity receptor, it strongly stimulates immunosuppressive CD4+ Tregs and seems to promote potentially life-threatening vascular leakage. IL-2/anti-IL-2 complexes (IL-2c), which bind to the low-affinity receptor, have been reported to circumvent these disadvantages but they have not yet been thoroughly tested in conjunction with radiotherapy.MethodsWe evaluated, in three mouse models, the antitumoral effects induced by hypofractionated RT (hRT) plus IL-2c. We also used non-invasive imaging with a newly developed PET tracer based on therapeutically active IL-2c and a PD-L1 PET tracer for the theranostic evaluation of the treatment and its side effects.ResultsTreatment of mice bearing established B16 melanomas with hRT + IL-2c was superior to hRT + uncomplexed IL-2 or hRT alone; IL-2c alone was not effective. hRT + IL-2c was also synergistic in mice bearing C51 colon carcinomas or 4T1 mammary carcinomas. The better antitumor response correlated with increased tumor-specific CD8+ T cells and NK cells, but not CD4+ Tregs, in the irradiated tumor and in lymphoid organs. With the new PET tracer, we visualized the whole-body distribution of IL-2c and the bound receptors in naïve mice and tumor-bearing mice. Surprisingly, the tumor uptake was non-specific and only moderate. This prompted experiments demonstrating that specific IL-2c binding in the tumor is limited by IL-2 secreted by tumor-resident effector cells and that extratumorally expanded T and NK cells can infiltrate the irradiated tumor, which suggests that systemic immune activation considerably contributed to the reduction of tumor growth. Lastly, we show that a side effect of IL-2c treatment – a quite dramatic non-specific expansion of CD8+ T and NK cells – is only transient, and we visualized the associated splenomegaly as well as side effects on liver and lung by contrast-enhanced CT and PD-L1 PET.ConclusionsOur results show that the combination of immunogenic RT with IL-2c that are directed towards the low-affinity IL-2 receptor can be synergistic and more effective than the combination with uncomplexed IL-2. In addition, our theranostic evaluation provided insights into the mechanism of action and the side effects of IL-2c treatment.

Highlights

  • Immunogenic radiotherapy (RT) can act synergistically with immune checkpoint blockers (ICBs)

  • IL-2 binds with lower affinity to the dimeric receptor consisting of CD122 and γc, which is expressed at high levels on memory CD8+ T cells and natural killer (NK) cells [3, 5]

  • When the tumors had reached approximately 250 mm3, mice were treated with IL-2 complexes (IL-2c) alone, hypofractionated RT (hRT) alone, hRT + IL-2, or hRT + IL-2c (Fig. 1b)

Read more

Summary

Introduction

Immunogenic radiotherapy (RT) can act synergistically with immune checkpoint blockers (ICBs). IL-2 has a short half-life, and, by binding to its high-affinity receptor, it strongly stimulates immunosuppressive CD4+ Tregs and seems to promote potentially life-threatening vascular leakage. The cytokine IL-2 is mainly secreted by effector T cells and is crucial for the proliferation, differentiation, and survival of T cells, including immunosuppressive CD4+ FoxP3+ regulatory T cells (Tregs). It is important for natural killer (NK) cells [1,2,3]. The pathophysiology of the IL-2-induced capillary leak syndrome seems multifactorial, binding of IL-2 to CD25 expressed on vascular endothelial cells has been reported to promote it [7, 8]

Methods
Results
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