Abstract

IL6 produced by tumor cells promotes their survival, conferring a poor prognosis in patients with cancer. IL6 also contributes to immunosuppression of CD4+ T cell-mediated antitumor effects. In this study, we focused on the impact of IL6 trans-signaling mediated by soluble IL6 receptors (sIL6R) expressed in tumor-bearing hosts. Higher levels of sIL6R circulating in blood were observed in tumor-bearing mice, whereas the systemic increase of sIL6R was not prominent in tumor-bearing mice with myeloid cell-specific conditional deletion of IL6R even when tumor cells produced sIL6R. Abundant sIL6R was released by CD11b+ cells from tumor-bearing mice but not tumor-free mice. Notably, IL6-mediated defects in Th1 differentiation, T-cell helper activity for tumor-specific CD8+ T cells, and downstream antitumor effects were rescued by myeloid-specific deletion of sIL6R. Expression of the T-cell transcription factor c-Maf was upregulated in CD4+ T cells primed in tumor-bearing mice in an IL6-dependent manner. Investigations with c-Maf loss-of-function T cells revealed that c-Maf activity was responsible for IL6/sIL6R-induced Th1 suppression and defective T-cell-mediated antitumor responses. In patients with cancer, myeloid cell-derived sIL6R was also possibly associated with Th1 suppression and c-Maf expression. Our results argued that increased expression of sIL6R from myeloid cells and subsequent c-Maf induction were adverse events for counteracting tumor-specific Th1 generation. Overall, this work provides a mechanistic rationale for sIL6R targeting to improve the efficacy of T-cell-mediated cancer immunotherapy. Cancer Res; 77(9); 2279-91. ©2017 AACR.

Highlights

  • The usage of T-cell–mediated cancer immunotherapies has gained momentum for achieving successes in advanced malignancies and is currently being hailed as a promising therapeutic approach [1, 2]

  • We previously demonstrated that differentiation of CD4þ T cells expressing OVA-specific T-cell receptor (TCR) (OT-II cells) into IFNg-producing Th1 cells was attenuated in mice with tumors expressing the surrogate tumor-associated antigen, OVA, and that the defect was reversed by anti-IL6 Ab treatment

  • This IL6-dependent Th1 suppression was mediated through the direct action of IL6 on CD4þ T cells because a substantial STAT3 activation was detected in donor OT-II cells in tumor-bearing mice, which was abrogated by in vivo administration of anti-IL6 receptor (IL6R) Ab (Fig. 1F)

Read more

Summary

Introduction

The usage of T-cell–mediated cancer immunotherapies has gained momentum for achieving successes in advanced malignancies and is currently being hailed as a promising therapeutic approach [1, 2]. While the induction of tumor-specific CD8þ T cells has been the main focus in cancer immunotherapy, recent studies have suggested that the better prognosis is correlated with higher levels of IFNg-expressing CD4þ T (Th1) cells in patients with cancer, implying an important role of Th1 cells in controlling. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

Methods
Results
Conclusion
Full Text
Paper version not known

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