Abstract

Immune checkpoint inhibitors that block the programmed cell death protein 1/PD-L1 pathway have significantly improved the survival of patients with advanced melanoma. Immunotherapies are only effective in 15–40% of melanoma patients and resistance is associated with defects in antigen presentation and interferon signaling pathways. In this study, we examined interferon-γ (IFNγ) responses in a large panel of immune checkpoint inhibitor-naïve melanoma cells with defined genetic drivers; BRAF-mutant (n = 11), NRAS-mutant (n = 10), BRAF/NRAS wild type (n = 10), and GNAQ/GNA11-mutant uveal melanomas (UVMs) (n = 8). Cell surface expression of established IFNγ downstream targets PD-L1, PD-L2, HLA-A, -B, and -C, HLA-DR, and nerve growth factor receptor (NGFR) were analyzed by flow cytometry. Basal cellular expression levels of HLA-A, -B, -C, HLA-DR, NGFR, and PD-L2 predicted the levels of IFNγ-stimulation, whereas PD-L1 induction was independent of basal expression levels. Only 13/39 (33%) of the melanoma cell lines tested responded to IFNγ with potent induction of all targets, indicating that downregulation of IFNγ signaling is common in melanoma. In addition, we identified two well-recognized mechanisms of immunotherapy resistance, the loss of β-2-microglobulin and interferon gamma receptor 1 expression. We also examined the influence of melanoma driver oncogenes on IFNγ signaling and our data suggest that UVM have diminished capacity to respond to IFNγ, with lower induced expression of several targets, consistent with the disappointing response of UVM to immunotherapies. Our results demonstrate that melanoma responses to IFNγ are heterogeneous, frequently downregulated in immune checkpoint inhibitor-naïve melanoma and potentially predictive of response to immunotherapy.

Highlights

  • The identification of checkpoint signaling pathways that dampen anti-cancer immune responses and the subsequent development of checkpoint inhibitors have transformed the treatment of patients with metastatic cancer

  • These cells were naïve to immune checkpoint inhibitors, and we examined whether the expression of key IFNγ downstream targets [PD-L1, PD-L2, nerve growth factor receptor (NGFR), HLA-A, -B, -C, and HLA-DR] could serve to assess the integrity of IFNγ signaling in melanoma

  • Analysis of the IFNγ target proteins, HLA-ABC, HLA-DR, NGFR, PD-L1, and PD-L2, in a panel of 39 melanoma cell lines revealed that IFNγ stimulated cell surface expression of all five markers in only 13 melanoma cell lines tested

Read more

Summary

Introduction

The identification of checkpoint signaling pathways that dampen anti-cancer immune responses and the subsequent development of checkpoint inhibitors have transformed the treatment of patients with metastatic cancer. Antibodies blocking immune checkpoints such as the cytotoxic T-lymphocyte-associated protein-4, the programmed cell death protein 1 (PD-1), and its ligand PD-L1 induce durable anti-tumor immune responses in many advanced malignancies, including melanoma, non-small-cell lung cancer, and renal cell carcinoma. Nuclear translocation of STAT transcription factors promotes the transcription of hundreds of IFNγ response genes [10] including downstream transcription factors, such as IRF1, STAT1, and STAT3, genes involved in antigen presentation such as MHC class I and II molecules [8, 11], and genes that attenuate immune activity to minimize local tissue damage, such as PD-L1 and PD-L2 [7]. The multifunctional effects of IFNγ are important in the context of immunotherapy since enhanced antigen presentation improves immune recognition of tumors while expression of immunosuppressive molecules limits anti-tumor T cell activity

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.