Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 8/2020

01-08-2020 | Glucocorticoid | Original Article

The glucocorticoids prednisone and dexamethasone differentially modulate T cell function in response to anti-PD-1 and anti-CTLA-4 immune checkpoint blockade

Authors: Isobel S. Okoye, Lai Xu, John Walker, Shokrollah Elahi

Published in: Cancer Immunology, Immunotherapy | Issue 8/2020

Login to get access

Abstract

On-treatment steroids for countering immune checkpoint inhibitor-induced inflammatory responses (irAEs) are a hallmark of cancer immunotherapy. However, the suppressive nature of steroids has raised questions regarding their ability to compromise the function of the ‘proliferative burst’ of effector T cells induced by immune checkpoint antibodies. We investigated the effector functions and the co-inhibitory receptor profile of stimulated peripheral blood mononuclear cells (PBMCs) pre-treated with prednisone and dexamethasone alone or in the presence of anti-PD-1/CTLA-4 antibodies. Also, clinical analysis of a patient who exhibited irAEs following combination (anti-PD-1/CTLA-4) in the presence of glucocorticoids was done. We found that prednisone in contrast to dexamethasone did not compromise T cell cytokine production (IL-2, IFN-γ and TNF-α) and proliferation in the absence or presence of anti-PD-1/CTLA-4 antibodies, when a physiological concentration was used. Neither single prednisone treatment nor co-treatment with checkpoint inhibitors impacted the expression of co-inhibitory receptors PD-1, CTLA-4, TIM-3 and LAG-3. In contrast, dexamethasone treatment promoted downregulation of LAG-3 expression by T cells. In addition, co-treatment of PD-1 + Jurkat cells with prednisone and/or dexamethasone with anti-PD-1 before stimulation significantly reduced SHP-2 phosphorylation, indicative of increased T cell function. Our findings hereby demonstrate a differential steroid effect on T cell function, which should be taken into consideration for patients undergoing immunotherapy. Also, the clinical analysis of a patient who exhibited irAEs following combination (anti-PD-1/CTLA-4) therapy indicated complete metabolic response in the presence of glucocorticoids. Therefore, concomitant use of prednisone does not appear to interfere with the function of immune checkpoint blockade.
Appendix
Available only for authorised users
Literature
2.
go back to reference Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DAA, Wherry EJ (2009) Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 10:29–37CrossRef Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DAA, Wherry EJ (2009) Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 10:29–37CrossRef
3.
go back to reference Thommen DS, Schreiner J, Muller P et al (2015) Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol Res 3(12):1344–1355CrossRef Thommen DS, Schreiner J, Muller P et al (2015) Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol Res 3(12):1344–1355CrossRef
4.
go back to reference Ribas A, Puzanov I, Dummer R et al (2015) Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 16:908–918CrossRef Ribas A, Puzanov I, Dummer R et al (2015) Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 16:908–918CrossRef
5.
go back to reference Callahan MK, Flaherty CR, Postow MA (2016) Checkpoint blockade for the treatment of advanced melanoma. Cancer Treat Res 167:231–250CrossRef Callahan MK, Flaherty CR, Postow MA (2016) Checkpoint blockade for the treatment of advanced melanoma. Cancer Treat Res 167:231–250CrossRef
12.
go back to reference Wherry EJ, Kurachi M (2015) Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 15:486–499CrossRef Wherry EJ, Kurachi M (2015) Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 15:486–499CrossRef
14.
go back to reference Michot JM, Bigenwald C, Champiat S et al (2016) Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur J Cancer 54:139–148CrossRef Michot JM, Bigenwald C, Champiat S et al (2016) Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur J Cancer 54:139–148CrossRef
17.
go back to reference Hofmann L, Forschner A, Loquai C et al (2016) Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer 60:190–209CrossRef Hofmann L, Forschner A, Loquai C et al (2016) Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer 60:190–209CrossRef
18.
go back to reference Wozniak S, Mackiewicz-Wysocka M, Krokowicz L, Kwinta L, Mackiewicz J (2015) Febrile neutropenia in a metastatic melanoma patient treated with ipilimumab—case report. Oncol Res Treat 38:105–108CrossRef Wozniak S, Mackiewicz-Wysocka M, Krokowicz L, Kwinta L, Mackiewicz J (2015) Febrile neutropenia in a metastatic melanoma patient treated with ipilimumab—case report. Oncol Res Treat 38:105–108CrossRef
19.
go back to reference Sibaud V, Meyer N, Lamant L, Vigarios E, Mazieres J, Delord JP (2016) Dermatologic complications of anti-PD-1/PD-L1 immune checkpoint antibodies. Curr Opin Oncol 28:254–263CrossRef Sibaud V, Meyer N, Lamant L, Vigarios E, Mazieres J, Delord JP (2016) Dermatologic complications of anti-PD-1/PD-L1 immune checkpoint antibodies. Curr Opin Oncol 28:254–263CrossRef
21.
go back to reference Horvat TZ, Adel NG, Dang T-O et al (2015) Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at Memorial Sloan Kettering Cancer Center. J Clin Oncol 33:3193–3198CrossRef Horvat TZ, Adel NG, Dang T-O et al (2015) Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at Memorial Sloan Kettering Cancer Center. J Clin Oncol 33:3193–3198CrossRef
24.
go back to reference Pawarode A, D’Souza A, Pasquini MC et al (2017) Phase 2 study of pembrolizumab during lymphodepleted state after autologous hematopoietic cell transplantation in multiple myeloma patients. Blood 130:339 Pawarode A, D’Souza A, Pasquini MC et al (2017) Phase 2 study of pembrolizumab during lymphodepleted state after autologous hematopoietic cell transplantation in multiple myeloma patients. Blood 130:339
26.
go back to reference Arbour KC, Mezquita L, Long N et al (2018) Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol 36:2872–2878CrossRef Arbour KC, Mezquita L, Long N et al (2018) Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J Clin Oncol 36:2872–2878CrossRef
32.
go back to reference Giles AJ, Hutchinson M-KND, Sonnemann HM et al (2018) Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer 6:51CrossRef Giles AJ, Hutchinson M-KND, Sonnemann HM et al (2018) Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer 6:51CrossRef
34.
go back to reference Okoye I, Namdar A, Xu L, Crux N, Elahi S (2017) Atorvastatin downregulates co-inhibitory receptor expression by targeting Ras-activated mTOR signalling. Oncotarget 8:98215–98232CrossRef Okoye I, Namdar A, Xu L, Crux N, Elahi S (2017) Atorvastatin downregulates co-inhibitory receptor expression by targeting Ras-activated mTOR signalling. Oncotarget 8:98215–98232CrossRef
35.
go back to reference Elahi S, Dinges WL, Lejarcegui N, Laing KJ, Collier AC, Koelle DM, McElrath MJ, Horton H (2011) Protective HIV-specific CD8(+) T cells evade T(reg) cell suppression. Nat Med 17:989–995CrossRef Elahi S, Dinges WL, Lejarcegui N, Laing KJ, Collier AC, Koelle DM, McElrath MJ, Horton H (2011) Protective HIV-specific CD8(+) T cells evade T(reg) cell suppression. Nat Med 17:989–995CrossRef
37.
go back to reference Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL (2004) SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol 173:945–954CrossRef Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL (2004) SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol 173:945–954CrossRef
48.
go back to reference Salmond RJ, Alexander DR (2006) SHP2 forecast for the immune system: fog gradually clearing. Trends Immunol 27:154–160CrossRef Salmond RJ, Alexander DR (2006) SHP2 forecast for the immune system: fog gradually clearing. Trends Immunol 27:154–160CrossRef
Metadata
Title
The glucocorticoids prednisone and dexamethasone differentially modulate T cell function in response to anti-PD-1 and anti-CTLA-4 immune checkpoint blockade
Authors
Isobel S. Okoye
Lai Xu
John Walker
Shokrollah Elahi
Publication date
01-08-2020
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 8/2020
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-020-02555-2

Other articles of this Issue 8/2020

Cancer Immunology, Immunotherapy 8/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine