Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Glucocorticoid-induced tumor necrosis factor receptor–related protein co-stimulation facilitates tumor regression by inducing IL-9–producing helper T cells

Abstract

T cell stimulation via glucocorticoid-induced tumor necrosis factor receptor (TNFR)-related protein (GITR) elicits antitumor activity in various tumor models; however, the underlying mechanism of action remains unclear. Here we demonstrate a crucial role for interleukin (IL)-9 in antitumor immunity generated by the GITR agonistic antibody DTA-1. IL-4 receptor knockout (Il4ra−/−) mice, which have reduced expression of IL-9, were resistant to tumor growth inhibition by DTA-1. Notably, neutralization of IL-9 considerably impaired tumor rejection induced by DTA-1. In particular, DTA-1–induced IL-9 promoted tumor-specific cytotoxic T lymphocyte (CTL) responses by enhancing the function of dendritic cells in vivo. Furthermore, GITR signaling enhanced the differentiation of IL-9–producing CD4+ T-helper (TH9) cells in a TNFR-associated factor 6 (TRAF6)- and NF-κB–dependent manner and inhibited the generation of induced regulatory T cells in vitro. Our findings demonstrate that GITR co-stimulation mediates antitumor immunity by promoting TH9 cell differentiation and enhancing CTL responses and thus provide a mechanism of action for GITR agonist–mediated cancer immunotherapies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: IL-4R signaling is essential for DTA-1–induced tumor growth inhibition.
Figure 2: DTA-1–mediated tumor regression requires IL-9.
Figure 3: GITR-triggered IL-9 production facilitates antitumor CTL responses by enhancing DC function in vivo.
Figure 4: GITR co-stimulation enhances mouse and human TH9 differentiation in vitro.
Figure 5: GITR triggering shifts the balance of iTreg cells to TH9 cells.
Figure 6: TRAF6–NF-κB pathway is associated with GITR-mediated TH9 differentiation.

Similar content being viewed by others

References

  1. Melero, I., Hervas-Stubbs, S., Glennie, M., Pardoll, D.M. & Chen, L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat. Rev. Cancer 7, 95–106 (2007).

    CAS  PubMed  Google Scholar 

  2. Peggs, K.S., Quezada, S.A., Korman, A.J. & Allison, J.P. Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Curr. Opin. Immunol. 18, 206–213 (2006).

    CAS  PubMed  Google Scholar 

  3. Maio, M., Di Giacomo, A.M., Robert, C. & Eggermont, A.M. Update on the role of ipilimumab in melanoma and first data on new combination therapies. Curr. Opin. Oncol. 25, 166–172 (2013).

    CAS  PubMed  Google Scholar 

  4. Topalian, S.L., Drake, C.G. & Pardoll, D.M. Targeting the PD-1–B7-H1(PD-L1) pathway to activate antitumor immunity. Curr. Opin. Immunol. 24, 207–212 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Tone, M. et al. Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells. Proc. Natl. Acad. Sci. USA 100, 15059–15064 (2003).

    CAS  PubMed  Google Scholar 

  6. Croft, M. Costimulatory members of the TNFR family: keys to effective T cell immunity? Nat. Rev. Immunol. 3, 609–620 (2003).

    CAS  PubMed  Google Scholar 

  7. Schaer, D.A., Murphy, J.T. & Wolchok, J.D. Modulation of GITR for cancer immunotherapy. Curr. Opin. Immunol. 24, 217–224 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhou, P., L'Italien, L., Hodges, D. & Schebye, X.M. Pivotal roles of CD4+ effector T cells in mediating agonistic anti-GITR mAb–induced immune activation and tumor immunity in CT26 tumors. J. Immunol. 179, 7365–7375 (2007).

    CAS  PubMed  Google Scholar 

  9. Ramirez-Montagut, T. et al. Glucocorticoid-induced TNF receptor family–related gene activation overcomes tolerance/ignorance to melanoma differentiation antigens and enhances antitumor immunity. J. Immunol. 176, 6434–6442 (2006).

    CAS  PubMed  Google Scholar 

  10. Nishikawa, H. et al. Regulatory T cell–resistant CD8+ T cells induced by glucocorticoid-induced tumor necrosis factor receptor signaling. Cancer Res. 68, 5948–5954 (2008).

    CAS  PubMed  Google Scholar 

  11. Zhou, P. et al. Mature B cells are critical to T cell–mediated tumor immunity induced by an agonist anti-GITR monoclonal antibody. J. Immunother. 33, 789–797 (2010).

    CAS  PubMed  Google Scholar 

  12. Ko, K. et al. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J. Exp. Med. 202, 885–891 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Cohen, A.D. et al. Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS ONE 5, e10436 (2010).

    PubMed  PubMed Central  Google Scholar 

  14. Shimizu, J., Yamazaki, S., Takahashi, T., Ishida, Y. & Sakaguchi, S. Stimulation of CD25+CD4+ regulatory T cells through GITR breaks immunological self-tolerance. Nat. Immunol. 3, 135–142 (2002).

    CAS  PubMed  Google Scholar 

  15. Muranski, P. & Restifo, N.P. Adoptive immunotherapy of cancer using CD4+ T cells. Curr. Opin. Immunol. 21, 200–208 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Dunn, G.P., Koebel, C.M. & Schreiber, R.D. Interferons, immunity and cancer immunoediting. Nat. Rev. Immunol. 6, 836–848 (2006).

    CAS  PubMed  Google Scholar 

  17. Kennedy, R. & Celis, E. Multiple roles for CD4+ T cells in antitumor immune responses. Immunol. Rev. 222, 129–144 (2008).

    CAS  PubMed  Google Scholar 

  18. Hong, S. et al. Roles of idiotype-specific T cells in myeloma cell growth and survival: TH1 and CTL cells are tumoricidal while TH2 cells promote tumor growth. Cancer Res. 68, 8456–8464 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. De Monte, L. et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J. Exp. Med. 208, 469–478 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Zou, W. Regulatory T cells, tumour immunity and immunotherapy. Nat. Rev. Immunol. 6, 295–307 (2006).

    CAS  PubMed  Google Scholar 

  21. Purwar, R. et al. Robust tumor immunity to melanoma mediated by interleukin-9–producing T cells. Nat. Med. 18, 1248–1253 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lu, Y. et al. TH9 cells promote antitumor immune responses in vivo. J. Clin. Invest. 122, 4160–4171 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Patel, M. et al. Glucocorticoid-induced TNFR family–related protein (GITR) activation exacerbates murine asthma and collagen-induced arthritis. Eur. J. Immunol. 35, 3581–3590 (2005).

    CAS  PubMed  Google Scholar 

  24. Motta, A.C. et al. GITR signaling potentiates airway hyperresponsiveness by enhancing TH2 cell activity in a mouse model of asthma. Respir. Res. 10, 93 (2009).

    PubMed  PubMed Central  Google Scholar 

  25. van der Werf, N. et al. TH2 responses to helminth parasites can be therapeutically enhanced by, but are not dependent upon, GITR–GITR ligand costimulation in vivo. J. Immunol. 187, 1411–1420 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhu, J., Yamane, H. & Paul, W.E. Differentiation of effector CD4 T cell populations. Annu. Rev. Immunol. 28, 445–489 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Veldhoen, M. et al. Transforming growth factor–β 'reprograms' the differentiation of T helper 2 cells and promotes an interleukin 9–producing subset. Nat. Immunol. 9, 1341–1346 (2008).

    CAS  PubMed  Google Scholar 

  28. Dardalhon, V. et al. IL-4 inhibits TGF-β–induced Foxp3+ T cells and, together with TGF-β, generates IL-9+IL-10+Foxp3 effector T cells. Nat. Immunol. 9, 1347–1355 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Végran, F. et al. The transcription factor IRF1 dictates the IL-21–dependent anticancer functions of TH9 cells. Nat. Immunol. 15, 758–766 (2014).

    PubMed  Google Scholar 

  30. Takami, M., Love, R.B. & Iwashima, M. TGF-β converts apoptotic stimuli into the signal for TH9 differentiation. J. Immunol. 188, 4369–4375 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Perumal, N.B. & Kaplan, M.H. Regulating Il9 transcription in T helper cells. Trends Immunol. 32, 146–150 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Chang, H.C. et al. The transcription factor PU.1 is required for the development of IL-9–producing T cells and allergic inflammation. Nat. Immunol. 11, 527–534 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Staudt, V. et al. Interferon-regulatory factor 4 is essential for the developmental program of T helper 9 cells. Immunity 33, 192–202 (2010).

    CAS  PubMed  Google Scholar 

  34. Jabeen, R. et al. TH9 cell development requires a BATF-regulated transcriptional network. J. Clin. Invest. 123, 4641–4653 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Stassen, M. et al. IL-9 and IL-13 production by activated mast cells is strongly enhanced in the presence of lipopolysaccharide: NF-κB is decisively involved in the expression of IL-9. J. Immunol. 166, 4391–4398 (2001).

    CAS  PubMed  Google Scholar 

  36. Jash, A. et al. Nuclear factor of activated T cells 1 (NFAT1)-induced permissive chromatin modification facilitates nuclear factor–kappaB (NF-kappaB)-mediated interleukin-9 (IL-9) transactivation. J. Biol. Chem. 287, 15445–15457 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Xiao, X. et al. OX40 signaling favors the induction of TH9 cells and airway inflammation. Nat. Immunol. 13, 981–990 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Ruffell, B., Affara, N.I. & Coussens, L.M. Differential macrophage programming in the tumor microenvironment. Trends Immunol. 33, 119–126 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Kitajima, M. et al. Memory type 2 helper T cells induce long-lasting antitumor immunity by activating natural killer cells. Cancer Res. 71, 4790–4798 (2011).

    CAS  PubMed  Google Scholar 

  40. Hung, K. et al. The central role of CD4+ T cells in the antitumor immune response. J. Exp. Med. 188, 2357–2368 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Mattes, J. et al. Immunotherapy of cytotoxic T cell–resistant tumors by T helper 2 cells: an eotaxin- and STAT6-dependent process. J. Exp. Med. 197, 387–393 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Stephens, G.L. et al. Engagement of glucocorticoid-induced TNFR family–related receptor on effector T cells by its ligand mediates resistance to suppression by CD4+CD25+ T cells. J. Immunol. 173, 5008–5020 (2004).

    CAS  PubMed  Google Scholar 

  43. Bulliard, Y. et al. Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J. Exp. Med. 210, 1685–1693 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Angkasekwinai, P., Chang, S.H., Thapa, M., Watarai, H. & Dong, C. Regulation of IL-9 expression by IL-25 signaling. Nat. Immunol. 11, 250–256 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Elyaman, W. et al. Notch receptors and Smad3 signaling cooperate in the induction of interleukin-9–producing T cells. Immunity 36, 623–634 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Kaplan, M.H. TH9 cells: differentiation and disease. Immunol. Rev. 252, 104–115 (2013).

    PubMed  PubMed Central  Google Scholar 

  47. Yao, W. et al. Interleukin-9 is required for allergic airway inflammation mediated by the cytokine TSLP. Immunity 38, 360–372 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Lin, G.H., Snell, L.M., Wortzman, M.E., Clouthier, D.L. & Watts, T.H. GITR-dependent regulation of 4-1BB expression: implications for T cell memory and anti–4-1BB–induced pathology. J. Immunol. 190, 4627–4639 (2013).

    CAS  PubMed  Google Scholar 

  49. Haribhai, D. et al. Regulatory T cells dynamically control the primary immune response to foreign antigen. J. Immunol. 178, 2961–2972 (2007).

    CAS  PubMed  Google Scholar 

  50. King, C.G. et al. TRAF6 is a T cell–intrinsic negative regulator required for the maintenance of immune homeostasis. Nat. Med. 12, 1088–1092 (2006).

    CAS  PubMed  Google Scholar 

  51. Vanneman, M. & Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 12, 237–251 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Johnson, L. et al. Somatic activation of the K-ras oncogene causes early-onset lung cancer in mice. Nature 410, 1111–1116 (2001).

    CAS  PubMed  Google Scholar 

  53. Kim, W.J. et al. Glucocorticoid-induced tumour necrosis factor receptor family related protein (GITR) mediates inflammatory activation of macrophages that can destabilize atherosclerotic plaques. Immunology 119, 421–429 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J.-O. Kim (International Vaccine Institute) for CD45.1 congenic mice, D.H. Chung (Seoul National University College of Medicine) for IL-4–deficient mice, Y.-K. Kim (Ewha Institute of Convergence Medicine) for IL-4Ra–deficient mice, T.H. Watts (University of Toronto) and P.P. Pandolfi (Harvard Medical School) for GITR-deficient mice, C.D. Surh (Pohang University of Science and Technology) and T.A. Chatila (Harvard Medical School) for Foxp3GFP reporter mice, Y. Choi (University of Pennsylvania School of Medicine) for Traf6fl/fl mice, G. Lozano (MD Anderson Cancer Center) for K-Ras transgenic mice, S. Sakaguchi (Osaka University) for the antibody to GITR (DTA-1), J.v. Snick (Ludwig Institute) for the antibody to IL-9 (MM9C1), S.-I. Nishikawa (RIKEN Center for Developmental Biology) for the antibody to c-kit (ACK2), H.-W. Yum (Seoul National University) and D.-H. Kim (Seoul National University) for assistance in setting up a colorectal cancer model, and members of the Kang laboratory for technical support. This work was supported by grants from the Public Welfare & Safety Research Program (20100020843; C.-Y.K.) and the National R&D Program for Cancer Control, Ministry of Health and Welfare (no. 0720500; C.-Y.K.).

Author information

Authors and Affiliations

Authors

Contributions

I.-K.K., B.-S.K., Y.C. and C.-Y.K. designed the study; I.-K.K., B.-S.K., C.-H.K., J.-W.S., J.-S.P., K.-S.S., E.-A.B., G.-E.L. and H.J. performed experiments; I.-K.K., B.-S.K., C.-H.K., J.-W.S., Y.C. and C.-Y.K. analyzed data; J.C., Y.J., D.H., B.S.K. and H.-Y.L. provided animals and reagents; I.-K.K., Y.C. and C.-Y.K. wrote the manuscript; C.-Y.K. supervised the study.

Corresponding author

Correspondence to Chang-Yuil Kang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and uncropped gels for western blots in main text. (PDF 3738 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, IK., Kim, BS., Koh, CH. et al. Glucocorticoid-induced tumor necrosis factor receptor–related protein co-stimulation facilitates tumor regression by inducing IL-9–producing helper T cells. Nat Med 21, 1010–1017 (2015). https://doi.org/10.1038/nm.3922

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3922

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer