Cargando…

Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy

PURPOSE: To investigate the impact of chemotherapy (CHT) on human retinoblastoma (RB) tumor microenvironment (TME). CASES AND METHODS: Ninety-four RBs were studied, including 44 primary RBs treated by upfront surgery (Group 1) and 50 primary RBs enucleated after CHT (CHT), either intra-arterial (IAC...

Descripción completa

Detalles Bibliográficos
Autores principales: Miracco, Clelia, Toti, Paolo, Gelmi, Maria Chiara, Aversa, Sara, Baldino, Gennaro, Galluzzi, Paolo, De Francesco, Sonia, Petrelli, Federica, Sorrentino, Ester, Belmonte, Giuseppe, Galimberti, Daniela, Bracco, Sandra, Hadjistilianou, Theodora
Formato: Online Artículo Texto
Lenguaje:English
Publicado: The Association for Research in Vision and Ophthalmology 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7862737/
https://www.ncbi.nlm.nih.gov/pubmed/33538768
http://dx.doi.org/10.1167/iovs.62.2.6
_version_ 1783647353890144256
author Miracco, Clelia
Toti, Paolo
Gelmi, Maria Chiara
Aversa, Sara
Baldino, Gennaro
Galluzzi, Paolo
De Francesco, Sonia
Petrelli, Federica
Sorrentino, Ester
Belmonte, Giuseppe
Galimberti, Daniela
Bracco, Sandra
Hadjistilianou, Theodora
author_facet Miracco, Clelia
Toti, Paolo
Gelmi, Maria Chiara
Aversa, Sara
Baldino, Gennaro
Galluzzi, Paolo
De Francesco, Sonia
Petrelli, Federica
Sorrentino, Ester
Belmonte, Giuseppe
Galimberti, Daniela
Bracco, Sandra
Hadjistilianou, Theodora
author_sort Miracco, Clelia
collection PubMed
description PURPOSE: To investigate the impact of chemotherapy (CHT) on human retinoblastoma (RB) tumor microenvironment (TME). CASES AND METHODS: Ninety-four RBs were studied, including 44 primary RBs treated by upfront surgery (Group 1) and 50 primary RBs enucleated after CHT (CHT), either intra-arterial (IAC; Group 2, 33 cases) or systemic (S-CHT; Group 3, 17 cases). Conventional and multiplexed immunohistochemistry were performed to make quantitative comparisons among the three groups, for the following parameters: tumor-infiltrating inflammatory cells (TI-ICs); programmed cell death protein 1 (PD-1) positive TI-ICs; Ki67 proliferation index; gliosis; PD-1 ligand (PD-L1) protein expression; vessel number. We also correlated these TME factors with the presence of histological high-risk factors (HHRF+) and RB anaplasia grade (AG). RESULTS: After CHT, a decrease in both RB burden and Ki67 positivity was observed. In parallel, most subsets of TI-ICs, PD-1+ TI-ICs, gliosis, and PD-L1 protein expression significantly increased (P < 0.001, P = 0.02, P < 0.001, respectively). Vessel number did not significantly vary. Age, HHRFs+ and AG were significantly different between primary and chemoreduced RBs (P < 0.001, P = 0.006, P = 0.001, respectively) and were correlated with most TME factors. CONCLUSIONS: CHT modulates host antitumor immunity by reorienting the RB TME from anergic into an active, CD8+, PD-L1+ hot state. Furthermore, some clinicopathological characteristics of RB correlate with several factors of TME. Our study adds data in favor of the possibility of a new therapeutic scenario in human RB.
format Online
Article
Text
id pubmed-7862737
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher The Association for Research in Vision and Ophthalmology
record_format MEDLINE/PubMed
spelling pubmed-78627372021-02-12 Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy Miracco, Clelia Toti, Paolo Gelmi, Maria Chiara Aversa, Sara Baldino, Gennaro Galluzzi, Paolo De Francesco, Sonia Petrelli, Federica Sorrentino, Ester Belmonte, Giuseppe Galimberti, Daniela Bracco, Sandra Hadjistilianou, Theodora Invest Ophthalmol Vis Sci Retina PURPOSE: To investigate the impact of chemotherapy (CHT) on human retinoblastoma (RB) tumor microenvironment (TME). CASES AND METHODS: Ninety-four RBs were studied, including 44 primary RBs treated by upfront surgery (Group 1) and 50 primary RBs enucleated after CHT (CHT), either intra-arterial (IAC; Group 2, 33 cases) or systemic (S-CHT; Group 3, 17 cases). Conventional and multiplexed immunohistochemistry were performed to make quantitative comparisons among the three groups, for the following parameters: tumor-infiltrating inflammatory cells (TI-ICs); programmed cell death protein 1 (PD-1) positive TI-ICs; Ki67 proliferation index; gliosis; PD-1 ligand (PD-L1) protein expression; vessel number. We also correlated these TME factors with the presence of histological high-risk factors (HHRF+) and RB anaplasia grade (AG). RESULTS: After CHT, a decrease in both RB burden and Ki67 positivity was observed. In parallel, most subsets of TI-ICs, PD-1+ TI-ICs, gliosis, and PD-L1 protein expression significantly increased (P < 0.001, P = 0.02, P < 0.001, respectively). Vessel number did not significantly vary. Age, HHRFs+ and AG were significantly different between primary and chemoreduced RBs (P < 0.001, P = 0.006, P = 0.001, respectively) and were correlated with most TME factors. CONCLUSIONS: CHT modulates host antitumor immunity by reorienting the RB TME from anergic into an active, CD8+, PD-L1+ hot state. Furthermore, some clinicopathological characteristics of RB correlate with several factors of TME. Our study adds data in favor of the possibility of a new therapeutic scenario in human RB. The Association for Research in Vision and Ophthalmology 2021-02-04 /pmc/articles/PMC7862737/ /pubmed/33538768 http://dx.doi.org/10.1167/iovs.62.2.6 Text en Copyright 2021 The Authors http://creativecommons.org/licenses/by-nc-nd/4.0/ This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.
spellingShingle Retina
Miracco, Clelia
Toti, Paolo
Gelmi, Maria Chiara
Aversa, Sara
Baldino, Gennaro
Galluzzi, Paolo
De Francesco, Sonia
Petrelli, Federica
Sorrentino, Ester
Belmonte, Giuseppe
Galimberti, Daniela
Bracco, Sandra
Hadjistilianou, Theodora
Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy
title Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy
title_full Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy
title_fullStr Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy
title_full_unstemmed Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy
title_short Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1− Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy
title_sort retinoblastoma is characterized by a cold, cd8+ cell poor, pd-l1− microenvironment, which turns into hot, cd8+ cell rich, pd-l1+ after chemotherapy
topic Retina
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7862737/
https://www.ncbi.nlm.nih.gov/pubmed/33538768
http://dx.doi.org/10.1167/iovs.62.2.6
work_keys_str_mv AT miraccoclelia retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT totipaolo retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT gelmimariachiara retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT aversasara retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT baldinogennaro retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT galluzzipaolo retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT defrancescosonia retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT petrellifederica retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT sorrentinoester retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT belmontegiuseppe retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT galimbertidaniela retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT braccosandra retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy
AT hadjistilianoutheodora retinoblastomaischaracterizedbyacoldcd8cellpoorpdl1microenvironmentwhichturnsintohotcd8cellrichpdl1afterchemotherapy