Programmed death-ligand-1 (PD-L1) plays a crucial role in the suppression of the antitumor immune response upon interaction with programmed cell death protein-1 (PD-1) on cytotoxic T cells. PD-L1 is constitutively expressed on antigen-presenting cells or tumor cells. Indeed, PD-L1 has been detected by immunohistochemistry in a variety of tumors, including malignant pleural mesothelioma (MPM), in which it has emerged as a predictive biomarker for PD-1/PD-L1 immune checkpoint blockers (ICBs).1,2
PD-L1 expression on tumor cells can be upregulated by several factors including the interferon gamma (IFN-γ) cytokine produced by tumor-infiltrating lymphocytes (TILs).3
In MPM cells, it has been reported that IFN-γ upregulates PD-L1 mRNA expression due to the activation of the interferon regulatory factor 1 (IRF1) transcription factor,4 but no data have been shown regarding cell surface PD-L1 that is functionally relevant for its contact with PD-1-positive cells.
Herein, we analyzed the ability of IFN-γ to upregulate the cell surface expression and release of PD-L1 in MPM cells. Both membranous and soluble PD-L1 may have implications in the tumor immune microenvironment and consequently in the clinical response of MPM to PD-1/PD-L1 ICBs.
Our study was conducted on the three MPM cell lines, MPP89, IST-MES1 and IST-MES2, all of which express a basal level of cell membranous PD-L1.5
Flow cytometry assessment showed that incubation of these cell lines with IFN-γ markedly increased membrane PD-L1 expression (by 4.02-, 7.76- and 11.40-fold in MPP89, IST-MES1 and IST-MES2, respectively) (Fig. 1a). This increase appeared to be dependent on the increase in the synthesis of PD-L1 protein, as shown by Western blot analysis of cell lysates (Fig. 1b). In turn, the synthesis of PD-L1 was associated with a strong IFN-γ-induced upregulation of the full-length transcript coding for membranous PD-L1 (mean expression fold change: 7.41, 21.61 and 15.84 in MPP89, IST-MES1 and IST-MES2 cells, respectively) (Fig. 1c). In addition, we found by ELISA that all cell lines were able to spontaneously secrete low amounts of the soluble form of PD-L1 (sPD-L1); more importantly, IFN-γ strongly increased the amount of sPD-L1 found in the culture supernatants (by 9.29-, 4.87- and 10.53-fold in MPP89, IST-MES1 and IST-MES2 cells, respectively) (Fig. 1d).
Thus, we provide the novel information that MPM cells can secrete sPD-L1 and that the levels of both membranous and soluble PD-L1 are upregulated by IFN-γ.
These findings may have a role in the clinical trials targeting either PD-1 or PD-L1 with ICBs that are currently underway in MPM in order to promote the antitumor immune response.2
In particular, since tumor cell-derived sPD-L1 is able to bind to the PD-1 receptor and to trigger pro-apoptotic signals into activated T cells,6 its binding to PD-1 may further contribute to an immunosuppressive microenvironment in MPM. Moreover, sPD-L1 might interfere with the efficacy of anti-PD-L1 antibodies by trapping them, as has recently been shown for circulating PD-L1 in non-small cell lung cancer patients.7 Consequently, sPD-L1 might also impair the anti-PD-L1 antibody-dependent cellular cytotoxicity (ADCC) of MPM cells induced by natural killer (NK) cells.8
In conclusion, we hypothesize that the high levels of IFN-γ that are released by TILs in MPM, via increased tumor cell surface expression and release of PD-L1, might contribute to the resistance to ICBs targeting PD-1 and/or PD-L1. Future investigations will be required to understand whether IFN-γ signaling blockade could increase the efficacy of immunotherapy with ICBs in MPM patients.
Acknowledgements
This work was supported by grants from the Italian Ministry of Health (5x1000 funds 2014 and 2015; Ricerca Corrente 2017).
Competing interests
The authors declare no competing interests.
References
- 1.Li X, Song W, Shao C, Shi Y, Han W. Emerging predictors of the response to the blockade of immune checkpoints in cancer therapy. Cell Mol. Immunol. 2019;16:28–39. doi: 10.1038/s41423-018-0086-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lantuejoul S, Le Stang N, Damiola F, Scherpereel A, Galateau-Sallé F. PD-L1 testing for immune checkpoint inhibitors in mesothelioma: for want of anything better? J. Thorac. Oncol. 2017;12:778–781. doi: 10.1016/j.jtho.2017.03.018. [DOI] [PubMed] [Google Scholar]
- 3.Mimura K, et al. PD-L1 expression is mainly regulated by interferon gamma associated with JAK-STAT pathway in gastric cancer. Cancer Sci. 2018;109:43–53. doi: 10.1111/cas.13424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kao SC, et al. Tumor suppressor microRNAs contribute to the regulation of PD-L1 expression in malignant pleural mesothelioma. J. Thorac. Oncol. 2017;12:1421–1433. doi: 10.1016/j.jtho.2017.05.024. [DOI] [PubMed] [Google Scholar]
- 5.Calabrò L, et al. CTLA4 blockade in mesothelioma: finally a competing strategy over cytotoxic/target therapy? Cancer Immunol. Immunother. 2015;64:105–112. doi: 10.1007/s00262-014-1609-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Frigola X, et al. Identification of a soluble form of B7-H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma. Clin. Cancer Res. 2011;17:1915–1923. doi: 10.1158/1078-0432.CCR-10-0250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gong B, et al. Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non-small cell lung cancer. J. Exp. Med. 2019;216:982–1000. doi: 10.1084/jem.20180870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Khanna S, et al. Malignant mesothelioma effusions are infiltrated by CD3+T cells highly expressing PD-L1 and the PD-L1+tumor cells within these effusions are susceptible to ADCC by the anti-PD-L1 antibody avelumab. J. Thorac. Oncol. 2016;11:1993–2005. doi: 10.1016/j.jtho.2016.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]