Cargando…

Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas

Tertiary lymphoid structures (TLS) are specialized lymphoid formations that serve as local repertoire of T- and B-cells at sites of chronic inflammation, autoimmunity, and cancer. While presence of TLS has been associated with improved response to immune checkpoint blockade therapies and overall out...

Descripción completa

Detalles Bibliográficos
Autores principales: Byers, Candice, Gill, Melissa, Kurtansky, Nicholas R., Alessi-Fox, Christi, Harman, Maggie, Cordova, Miguel, Gonzalez, Salvador, Guitera, Pascale, Rotemberg, Veronica, Marghoob, Ashfaq, Chen, Chih-Shan Jason, Dy, Jennifer, Kose, Kivanc, Rajadhyaksha, Milind, Sahu, Aditi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9647637/
https://www.ncbi.nlm.nih.gov/pubmed/36388913
http://dx.doi.org/10.3389/fmed.2022.981074
_version_ 1784827417773211648
author Byers, Candice
Gill, Melissa
Kurtansky, Nicholas R.
Alessi-Fox, Christi
Harman, Maggie
Cordova, Miguel
Gonzalez, Salvador
Guitera, Pascale
Rotemberg, Veronica
Marghoob, Ashfaq
Chen, Chih-Shan Jason
Dy, Jennifer
Kose, Kivanc
Rajadhyaksha, Milind
Sahu, Aditi
author_facet Byers, Candice
Gill, Melissa
Kurtansky, Nicholas R.
Alessi-Fox, Christi
Harman, Maggie
Cordova, Miguel
Gonzalez, Salvador
Guitera, Pascale
Rotemberg, Veronica
Marghoob, Ashfaq
Chen, Chih-Shan Jason
Dy, Jennifer
Kose, Kivanc
Rajadhyaksha, Milind
Sahu, Aditi
author_sort Byers, Candice
collection PubMed
description Tertiary lymphoid structures (TLS) are specialized lymphoid formations that serve as local repertoire of T- and B-cells at sites of chronic inflammation, autoimmunity, and cancer. While presence of TLS has been associated with improved response to immune checkpoint blockade therapies and overall outcomes in several cancers, its prognostic value in basal cell carcinoma (BCC) has not been investigated. Herein, we determined the prognostic impact of TLS by relating its prevalence and maturation with outcome measures of anti-tumor immunity, namely tumor infiltrating lymphocytes (TILs) and tumor killing. In 30 distinct BCCs, we show the presence of TLS was significantly enriched in tumors harboring a nodular component and more mature primary TLS was associated with TIL counts. Moreover, assessment of the fibrillary matrix surrounding tumors showed discrete morphologies significantly associated with higher TIL counts, critically accounting for heterogeneity in TIL count distribution within TLS maturation stages. Specifically, increased length of fibers and lacunarity of the matrix with concomitant reduction in density and alignment of fibers were present surrounding tumors displaying high TIL counts. Given the interest in inducing TLS formation as a therapeutic intervention as well as its documented prognostic value, elucidating potential impediments to the ability of TLS in driving anti-tumor immunity within the tumor microenvironment warrants further investigation. These results begin to address and highlight the need to integrate stromal features which may present a hindrance to TLS formation and/or effective function as a mediator of immunotherapy response.
format Online
Article
Text
id pubmed-9647637
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-96476372022-11-15 Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas Byers, Candice Gill, Melissa Kurtansky, Nicholas R. Alessi-Fox, Christi Harman, Maggie Cordova, Miguel Gonzalez, Salvador Guitera, Pascale Rotemberg, Veronica Marghoob, Ashfaq Chen, Chih-Shan Jason Dy, Jennifer Kose, Kivanc Rajadhyaksha, Milind Sahu, Aditi Front Med (Lausanne) Medicine Tertiary lymphoid structures (TLS) are specialized lymphoid formations that serve as local repertoire of T- and B-cells at sites of chronic inflammation, autoimmunity, and cancer. While presence of TLS has been associated with improved response to immune checkpoint blockade therapies and overall outcomes in several cancers, its prognostic value in basal cell carcinoma (BCC) has not been investigated. Herein, we determined the prognostic impact of TLS by relating its prevalence and maturation with outcome measures of anti-tumor immunity, namely tumor infiltrating lymphocytes (TILs) and tumor killing. In 30 distinct BCCs, we show the presence of TLS was significantly enriched in tumors harboring a nodular component and more mature primary TLS was associated with TIL counts. Moreover, assessment of the fibrillary matrix surrounding tumors showed discrete morphologies significantly associated with higher TIL counts, critically accounting for heterogeneity in TIL count distribution within TLS maturation stages. Specifically, increased length of fibers and lacunarity of the matrix with concomitant reduction in density and alignment of fibers were present surrounding tumors displaying high TIL counts. Given the interest in inducing TLS formation as a therapeutic intervention as well as its documented prognostic value, elucidating potential impediments to the ability of TLS in driving anti-tumor immunity within the tumor microenvironment warrants further investigation. These results begin to address and highlight the need to integrate stromal features which may present a hindrance to TLS formation and/or effective function as a mediator of immunotherapy response. Frontiers Media S.A. 2022-10-27 /pmc/articles/PMC9647637/ /pubmed/36388913 http://dx.doi.org/10.3389/fmed.2022.981074 Text en Copyright © 2022 Byers, Gill, Kurtansky, Alessi-Fox, Harman, Cordova, Gonzalez, Guitera, Rotemberg, Marghoob, Chen, Dy, Kose, Rajadhyaksha and Sahu. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Medicine
Byers, Candice
Gill, Melissa
Kurtansky, Nicholas R.
Alessi-Fox, Christi
Harman, Maggie
Cordova, Miguel
Gonzalez, Salvador
Guitera, Pascale
Rotemberg, Veronica
Marghoob, Ashfaq
Chen, Chih-Shan Jason
Dy, Jennifer
Kose, Kivanc
Rajadhyaksha, Milind
Sahu, Aditi
Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas
title Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas
title_full Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas
title_fullStr Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas
title_full_unstemmed Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas
title_short Tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas
title_sort tertiary lymphoid structures accompanied by fibrillary matrix morphology impact anti-tumor immunity in basal cell carcinomas
topic Medicine
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9647637/
https://www.ncbi.nlm.nih.gov/pubmed/36388913
http://dx.doi.org/10.3389/fmed.2022.981074
work_keys_str_mv AT byerscandice tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT gillmelissa tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT kurtanskynicholasr tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT alessifoxchristi tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT harmanmaggie tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT cordovamiguel tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT gonzalezsalvador tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT guiterapascale tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT rotembergveronica tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT marghoobashfaq tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT chenchihshanjason tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT dyjennifer tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT kosekivanc tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT rajadhyakshamilind tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas
AT sahuaditi tertiarylymphoidstructuresaccompaniedbyfibrillarymatrixmorphologyimpactantitumorimmunityinbasalcellcarcinomas