Cargando…

Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1

Adoptive immunotherapy is based on ex vivo expansion and stimulation of T-cells, followed by their transfer into patients. The need for the ex vivo culturing step provides opportunities for modulating the properties of transferred T-cells, enhancing their antitumor abilities, and increasing their nu...

Descripción completa

Detalles Bibliográficos
Autores principales: Adutler-Lieber, Shimrit, Friedman, Nir, Geiger, Benjamin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6004589/
https://www.ncbi.nlm.nih.gov/pubmed/29942308
http://dx.doi.org/10.3389/fimmu.2018.01303
_version_ 1783332546741796864
author Adutler-Lieber, Shimrit
Friedman, Nir
Geiger, Benjamin
author_facet Adutler-Lieber, Shimrit
Friedman, Nir
Geiger, Benjamin
author_sort Adutler-Lieber, Shimrit
collection PubMed
description Adoptive immunotherapy is based on ex vivo expansion and stimulation of T-cells, followed by their transfer into patients. The need for the ex vivo culturing step provides opportunities for modulating the properties of transferred T-cells, enhancing their antitumor abilities, and increasing their number. Here, we present a synthetic immune niche (SIN) that increases the number and antitumor activity of cytotoxic CD8(+) T-cells. We first evaluated the effect of various SIN compositions that mimic the physiological microenvironment encountered by T-cells during their activation and expansion in the lymph node. We found that substrates coated with the chemokine CCL21 together with the adhesion molecule intercellular adhesion molecule 1 significantly increase the number of ovalbumin-specific murine CD8(+) T-cells activated by antigen-loaded dendritic cells or activation microbeads. Notably, cells cultured on these substrates also displayed augmented cytotoxic activity toward ovalbumin-expressing melanoma cells, both in culture and in vivo. This increase in specific cytotoxic activity was associated with a major increase in the cellular levels of the killing-mediator granzyme B. Our results suggest that this SIN may be used for generating T-cells with augmented cytotoxic function, for use in cancer immunotherapy.
format Online
Article
Text
id pubmed-6004589
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-60045892018-06-25 Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1 Adutler-Lieber, Shimrit Friedman, Nir Geiger, Benjamin Front Immunol Immunology Adoptive immunotherapy is based on ex vivo expansion and stimulation of T-cells, followed by their transfer into patients. The need for the ex vivo culturing step provides opportunities for modulating the properties of transferred T-cells, enhancing their antitumor abilities, and increasing their number. Here, we present a synthetic immune niche (SIN) that increases the number and antitumor activity of cytotoxic CD8(+) T-cells. We first evaluated the effect of various SIN compositions that mimic the physiological microenvironment encountered by T-cells during their activation and expansion in the lymph node. We found that substrates coated with the chemokine CCL21 together with the adhesion molecule intercellular adhesion molecule 1 significantly increase the number of ovalbumin-specific murine CD8(+) T-cells activated by antigen-loaded dendritic cells or activation microbeads. Notably, cells cultured on these substrates also displayed augmented cytotoxic activity toward ovalbumin-expressing melanoma cells, both in culture and in vivo. This increase in specific cytotoxic activity was associated with a major increase in the cellular levels of the killing-mediator granzyme B. Our results suggest that this SIN may be used for generating T-cells with augmented cytotoxic function, for use in cancer immunotherapy. Frontiers Media S.A. 2018-06-11 /pmc/articles/PMC6004589/ /pubmed/29942308 http://dx.doi.org/10.3389/fimmu.2018.01303 Text en Copyright © 2018 Adutler-Lieber, Friedman and Geiger. 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 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 Immunology
Adutler-Lieber, Shimrit
Friedman, Nir
Geiger, Benjamin
Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1
title Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1
title_full Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1
title_fullStr Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1
title_full_unstemmed Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1
title_short Expansion and Antitumor Cytotoxicity of T-Cells Are Augmented by Substrate-Bound CCL21 and Intercellular Adhesion Molecule 1
title_sort expansion and antitumor cytotoxicity of t-cells are augmented by substrate-bound ccl21 and intercellular adhesion molecule 1
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6004589/
https://www.ncbi.nlm.nih.gov/pubmed/29942308
http://dx.doi.org/10.3389/fimmu.2018.01303
work_keys_str_mv AT adutlerliebershimrit expansionandantitumorcytotoxicityoftcellsareaugmentedbysubstrateboundccl21andintercellularadhesionmolecule1
AT friedmannir expansionandantitumorcytotoxicityoftcellsareaugmentedbysubstrateboundccl21andintercellularadhesionmolecule1
AT geigerbenjamin expansionandantitumorcytotoxicityoftcellsareaugmentedbysubstrateboundccl21andintercellularadhesionmolecule1