Cargando…
Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma
BACKGROUND: Immune responses against tumors are subject to negative feedback regulation. Immune checkpoint inhibitors (ICIs) blocking Programmed cell death protein 1 (PD-1), a receptor expressed on T cells, or its ligand PD-L1 have significantly improved the treatment of cancer, in particular malign...
Autores principales: | , , , , , , , , , , , , , , , , , , , , , , , , , , , |
---|---|
Formato: | Online Artículo Texto |
Lenguaje: | English |
Publicado: |
BMJ Publishing Group
2023
|
Materias: | |
Acceso en línea: | https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10083881/ https://www.ncbi.nlm.nih.gov/pubmed/37028818 http://dx.doi.org/10.1136/jitc-2021-004150 |
_version_ | 1785021617918705664 |
---|---|
author | Martins Nascentes Melo, Luiza Herrera-Rios, Dayana Hinze, Daniel Löffek, Stefanie Oezel, Irem Turiello, Roberta Klein, Juliane Leonardelli, Sonia Westedt, Isa-Vanessa Al-Matary, Yahya Egea-Rodriguez, Sara Brenzel, Alexandra Bau, Maja Sucker, Antje Hadaschik, Eva Wirsdörfer, Florian Hanenberg, Helmut Uhlenbrock, Niklas Rauh, Daniel Poźniak, Joanna Rambow, Florian Marine, Jean-Christophe Effern, Maike Glodde, Nicole Schadendorf, Dirk Jablonska, Jadwiga Hölzel, Michael Helfrich, Iris |
author_facet | Martins Nascentes Melo, Luiza Herrera-Rios, Dayana Hinze, Daniel Löffek, Stefanie Oezel, Irem Turiello, Roberta Klein, Juliane Leonardelli, Sonia Westedt, Isa-Vanessa Al-Matary, Yahya Egea-Rodriguez, Sara Brenzel, Alexandra Bau, Maja Sucker, Antje Hadaschik, Eva Wirsdörfer, Florian Hanenberg, Helmut Uhlenbrock, Niklas Rauh, Daniel Poźniak, Joanna Rambow, Florian Marine, Jean-Christophe Effern, Maike Glodde, Nicole Schadendorf, Dirk Jablonska, Jadwiga Hölzel, Michael Helfrich, Iris |
author_sort | Martins Nascentes Melo, Luiza |
collection | PubMed |
description | BACKGROUND: Immune responses against tumors are subject to negative feedback regulation. Immune checkpoint inhibitors (ICIs) blocking Programmed cell death protein 1 (PD-1), a receptor expressed on T cells, or its ligand PD-L1 have significantly improved the treatment of cancer, in particular malignant melanoma. Nevertheless, responses and durability are variables, suggesting that additional critical negative feedback mechanisms exist and need to be targeted to improve therapeutic efficacy. METHODS: We used different syngeneic melanoma mouse models and performed PD-1 blockade to identify novel mechanisms of negative immune regulation. Genetic gain-of-function and loss-of-function approaches as well as small molecule inhibitor applications were used for target validation in our melanoma models. We analyzed mouse melanoma tissues from treated and untreated mice by RNA-seq, immunofluorescence and flow cytometry to detect changes in pathway activities and immune cell composition of the tumor microenvironment. We analyzed tissue sections of patients with melanoma by immunohistochemistry as well as publicly available single-cell RNA-seq data and correlated target expression with clinical responses to ICIs. RESULTS: Here, we identified 11-beta-hydroxysteroid dehydrogenase-1 (HSD11B1), an enzyme that converts inert glucocorticoids into active forms in tissues, as negative feedback mechanism in response to T cell immunotherapies. Glucocorticoids are potent suppressors of immune responses. HSD11B1 was expressed in different cellular compartments of melanomas, most notably myeloid cells but also T cells and melanoma cells. Enforced expression of HSD11B1 in mouse melanomas limited the efficacy of PD-1 blockade, whereas small molecule HSD11B1 inhibitors improved responses in a CD8(+) T cell-dependent manner. Mechanistically, HSD11B1 inhibition in combination with PD-1 blockade augmented the production of interferon-γ by T cells. Interferon pathway activation correlated with sensitivity to PD-1 blockade linked to anti-proliferative effects on melanoma cells. Furthermore, high levels of HSD11B1, predominantly expressed by tumor-associated macrophages, were associated with poor responses to ICI therapy in two independent cohorts of patients with advanced melanomas analyzed by different methods (scRNA-seq, immunohistochemistry). CONCLUSION: As HSD11B1 inhibitors are in the focus of drug development for metabolic diseases, our data suggest a drug repurposing strategy combining HSD11B1 inhibitors with ICIs to improve melanoma immunotherapy. Furthermore, our work also delineated potential caveats emphasizing the need for careful patient stratification. |
format | Online Article Text |
id | pubmed-10083881 |
institution | National Center for Biotechnology Information |
language | English |
publishDate | 2023 |
publisher | BMJ Publishing Group |
record_format | MEDLINE/PubMed |
spelling | pubmed-100838812023-04-11 Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma Martins Nascentes Melo, Luiza Herrera-Rios, Dayana Hinze, Daniel Löffek, Stefanie Oezel, Irem Turiello, Roberta Klein, Juliane Leonardelli, Sonia Westedt, Isa-Vanessa Al-Matary, Yahya Egea-Rodriguez, Sara Brenzel, Alexandra Bau, Maja Sucker, Antje Hadaschik, Eva Wirsdörfer, Florian Hanenberg, Helmut Uhlenbrock, Niklas Rauh, Daniel Poźniak, Joanna Rambow, Florian Marine, Jean-Christophe Effern, Maike Glodde, Nicole Schadendorf, Dirk Jablonska, Jadwiga Hölzel, Michael Helfrich, Iris J Immunother Cancer Basic Tumor Immunology BACKGROUND: Immune responses against tumors are subject to negative feedback regulation. Immune checkpoint inhibitors (ICIs) blocking Programmed cell death protein 1 (PD-1), a receptor expressed on T cells, or its ligand PD-L1 have significantly improved the treatment of cancer, in particular malignant melanoma. Nevertheless, responses and durability are variables, suggesting that additional critical negative feedback mechanisms exist and need to be targeted to improve therapeutic efficacy. METHODS: We used different syngeneic melanoma mouse models and performed PD-1 blockade to identify novel mechanisms of negative immune regulation. Genetic gain-of-function and loss-of-function approaches as well as small molecule inhibitor applications were used for target validation in our melanoma models. We analyzed mouse melanoma tissues from treated and untreated mice by RNA-seq, immunofluorescence and flow cytometry to detect changes in pathway activities and immune cell composition of the tumor microenvironment. We analyzed tissue sections of patients with melanoma by immunohistochemistry as well as publicly available single-cell RNA-seq data and correlated target expression with clinical responses to ICIs. RESULTS: Here, we identified 11-beta-hydroxysteroid dehydrogenase-1 (HSD11B1), an enzyme that converts inert glucocorticoids into active forms in tissues, as negative feedback mechanism in response to T cell immunotherapies. Glucocorticoids are potent suppressors of immune responses. HSD11B1 was expressed in different cellular compartments of melanomas, most notably myeloid cells but also T cells and melanoma cells. Enforced expression of HSD11B1 in mouse melanomas limited the efficacy of PD-1 blockade, whereas small molecule HSD11B1 inhibitors improved responses in a CD8(+) T cell-dependent manner. Mechanistically, HSD11B1 inhibition in combination with PD-1 blockade augmented the production of interferon-γ by T cells. Interferon pathway activation correlated with sensitivity to PD-1 blockade linked to anti-proliferative effects on melanoma cells. Furthermore, high levels of HSD11B1, predominantly expressed by tumor-associated macrophages, were associated with poor responses to ICI therapy in two independent cohorts of patients with advanced melanomas analyzed by different methods (scRNA-seq, immunohistochemistry). CONCLUSION: As HSD11B1 inhibitors are in the focus of drug development for metabolic diseases, our data suggest a drug repurposing strategy combining HSD11B1 inhibitors with ICIs to improve melanoma immunotherapy. Furthermore, our work also delineated potential caveats emphasizing the need for careful patient stratification. BMJ Publishing Group 2023-04-07 /pmc/articles/PMC10083881/ /pubmed/37028818 http://dx.doi.org/10.1136/jitc-2021-004150 Text en © Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) . |
spellingShingle | Basic Tumor Immunology Martins Nascentes Melo, Luiza Herrera-Rios, Dayana Hinze, Daniel Löffek, Stefanie Oezel, Irem Turiello, Roberta Klein, Juliane Leonardelli, Sonia Westedt, Isa-Vanessa Al-Matary, Yahya Egea-Rodriguez, Sara Brenzel, Alexandra Bau, Maja Sucker, Antje Hadaschik, Eva Wirsdörfer, Florian Hanenberg, Helmut Uhlenbrock, Niklas Rauh, Daniel Poźniak, Joanna Rambow, Florian Marine, Jean-Christophe Effern, Maike Glodde, Nicole Schadendorf, Dirk Jablonska, Jadwiga Hölzel, Michael Helfrich, Iris Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma |
title | Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma |
title_full | Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma |
title_fullStr | Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma |
title_full_unstemmed | Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma |
title_short | Glucocorticoid activation by HSD11B1 limits T cell-driven interferon signaling and response to PD-1 blockade in melanoma |
title_sort | glucocorticoid activation by hsd11b1 limits t cell-driven interferon signaling and response to pd-1 blockade in melanoma |
topic | Basic Tumor Immunology |
url | https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10083881/ https://www.ncbi.nlm.nih.gov/pubmed/37028818 http://dx.doi.org/10.1136/jitc-2021-004150 |
work_keys_str_mv | AT martinsnascentesmeloluiza glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT herrerariosdayana glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT hinzedaniel glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT loffekstefanie glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT oezelirem glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT turielloroberta glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT kleinjuliane glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT leonardellisonia glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT westedtisavanessa glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT almataryyahya glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT egearodriguezsara glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT brenzelalexandra glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT baumaja glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT suckerantje glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT hadaschikeva glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT wirsdorferflorian glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT hanenberghelmut glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT uhlenbrockniklas glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT rauhdaniel glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT pozniakjoanna glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT rambowflorian glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT marinejeanchristophe glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT effernmaike glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT gloddenicole glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT schadendorfdirk glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT jablonskajadwiga glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT holzelmichael glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma AT helfrichiris glucocorticoidactivationbyhsd11b1limitstcelldriveninterferonsignalingandresponsetopd1blockadeinmelanoma |