Cargando…

FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation

Uterine sarcomas are rare but deadly malignancies without effective treatment. Immunotherapy is a promising new approach to treat these tumors but has shown heterogeneous effects in sarcoma patients. With the goal of identifying key factors for improved patient treatment, we characterized the tumor...

Descripción completa

Detalles Bibliográficos
Autores principales: Gultekin, Okan, Gonzalez-Molina, Jordi, Hardell, Elin, Moyano-Galceran, Lidia, Mitsios, Nicholas, Mulder, Jan, Kokaraki, Georgia, Isaksson, Anders, Sarhan, Dhifaf, Lehti, Kaisa, Carlson, Joseph W.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8604926/
https://www.ncbi.nlm.nih.gov/pubmed/34799669
http://dx.doi.org/10.1038/s41698-021-00236-6
_version_ 1784602064193585152
author Gultekin, Okan
Gonzalez-Molina, Jordi
Hardell, Elin
Moyano-Galceran, Lidia
Mitsios, Nicholas
Mulder, Jan
Kokaraki, Georgia
Isaksson, Anders
Sarhan, Dhifaf
Lehti, Kaisa
Carlson, Joseph W.
author_facet Gultekin, Okan
Gonzalez-Molina, Jordi
Hardell, Elin
Moyano-Galceran, Lidia
Mitsios, Nicholas
Mulder, Jan
Kokaraki, Georgia
Isaksson, Anders
Sarhan, Dhifaf
Lehti, Kaisa
Carlson, Joseph W.
author_sort Gultekin, Okan
collection PubMed
description Uterine sarcomas are rare but deadly malignancies without effective treatment. Immunotherapy is a promising new approach to treat these tumors but has shown heterogeneous effects in sarcoma patients. With the goal of identifying key factors for improved patient treatment, we characterized the tumor immune landscape in 58 uterine sarcoma cases with full clinicopathological annotation. Immune cell characterization revealed the overall prevalence of FOXP3+ cells and pro-tumor M2-like macrophages. Hierarchical clustering of patients showed four tumor type-independent immune signatures, where infiltration of FOXP3+ cells and M1-like macrophages associated with favorable prognosis. High CD8+/FOXP3+ ratio in UUS and ESS correlated with poor survival, upregulation of immunosuppressive markers, extracellular matrix (ECM)-related genes and proteins, and YAP activation. This study shows that uterine sarcomas present distinct immune signatures with prognostic value, independent of tumor type, and suggests that targeting the ECM could be beneficial for future treatments.
format Online
Article
Text
id pubmed-8604926
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-86049262021-12-03 FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation Gultekin, Okan Gonzalez-Molina, Jordi Hardell, Elin Moyano-Galceran, Lidia Mitsios, Nicholas Mulder, Jan Kokaraki, Georgia Isaksson, Anders Sarhan, Dhifaf Lehti, Kaisa Carlson, Joseph W. NPJ Precis Oncol Article Uterine sarcomas are rare but deadly malignancies without effective treatment. Immunotherapy is a promising new approach to treat these tumors but has shown heterogeneous effects in sarcoma patients. With the goal of identifying key factors for improved patient treatment, we characterized the tumor immune landscape in 58 uterine sarcoma cases with full clinicopathological annotation. Immune cell characterization revealed the overall prevalence of FOXP3+ cells and pro-tumor M2-like macrophages. Hierarchical clustering of patients showed four tumor type-independent immune signatures, where infiltration of FOXP3+ cells and M1-like macrophages associated with favorable prognosis. High CD8+/FOXP3+ ratio in UUS and ESS correlated with poor survival, upregulation of immunosuppressive markers, extracellular matrix (ECM)-related genes and proteins, and YAP activation. This study shows that uterine sarcomas present distinct immune signatures with prognostic value, independent of tumor type, and suggests that targeting the ECM could be beneficial for future treatments. Nature Publishing Group UK 2021-11-19 /pmc/articles/PMC8604926/ /pubmed/34799669 http://dx.doi.org/10.1038/s41698-021-00236-6 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Gultekin, Okan
Gonzalez-Molina, Jordi
Hardell, Elin
Moyano-Galceran, Lidia
Mitsios, Nicholas
Mulder, Jan
Kokaraki, Georgia
Isaksson, Anders
Sarhan, Dhifaf
Lehti, Kaisa
Carlson, Joseph W.
FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation
title FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation
title_full FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation
title_fullStr FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation
title_full_unstemmed FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation
title_short FOXP3+ T cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced YAP activation
title_sort foxp3+ t cells in uterine sarcomas are associated with favorable prognosis, low extracellular matrix expression and reduced yap activation
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8604926/
https://www.ncbi.nlm.nih.gov/pubmed/34799669
http://dx.doi.org/10.1038/s41698-021-00236-6
work_keys_str_mv AT gultekinokan foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT gonzalezmolinajordi foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT hardellelin foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT moyanogalceranlidia foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT mitsiosnicholas foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT mulderjan foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT kokarakigeorgia foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT isakssonanders foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT sarhandhifaf foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT lehtikaisa foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation
AT carlsonjosephw foxp3tcellsinuterinesarcomasareassociatedwithfavorableprognosislowextracellularmatrixexpressionandreducedyapactivation