Cargando…

Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens

Monocyte-derived Dendritic cells (DCs) have successfully been employed to induce immune responses against tumor-associated antigens in patients with various cancer entities. However, objective clinical responses have only been achieved in a minority of patients. Additionally, generation of GMP-compl...

Descripción completa

Detalles Bibliográficos
Autores principales: Hänel, Gerulf, Angerer, Caroline, Petry, Katja, Lichtenegger, Felix S., Subklewe, Marion
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer Berlin Heidelberg 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8614222/
https://www.ncbi.nlm.nih.gov/pubmed/34821951
http://dx.doi.org/10.1007/s00262-021-03109-w
_version_ 1784603813705940992
author Hänel, Gerulf
Angerer, Caroline
Petry, Katja
Lichtenegger, Felix S.
Subklewe, Marion
author_facet Hänel, Gerulf
Angerer, Caroline
Petry, Katja
Lichtenegger, Felix S.
Subklewe, Marion
author_sort Hänel, Gerulf
collection PubMed
description Monocyte-derived Dendritic cells (DCs) have successfully been employed to induce immune responses against tumor-associated antigens in patients with various cancer entities. However, objective clinical responses have only been achieved in a minority of patients. Additionally, generation of GMP-compliant DCs requires time- and labor-intensive cell differentiation. In contrast, Blood DCs (BDCs) require only minimal ex vivo handling, as differentiation occurs in vivo resulting in potentially better functional capacities and survival. We aimed to identify a protocol for optimal in vitro activation of BDCs including the three subsets pDCs, cDC1s, and cDC2s. We evaluated several TLR ligand combinations and demonstrated that polyinosinic:polycytidylic acid [poly(I:C)] and R848, ligands for TLR3 and TLR7/8, respectively, constituted the optimal combination for inducing a positive co-stimulatory profile in all BDC subsets. In addition, TLR3 and TLR7/8 activation led to high secretion of IFN-α and IL-12p70. Simultaneous as opposed to separate tailored activation of pDCs and cDCs increased immunostimulatory capacities, suggesting that BDC subsets engage in synergistic cross-talk during activation. Stimulation of BDCs with this protocol resulted in enhanced migration, high NK-cell activation, and potent antigen-specific T-cell induction. We conclude that simultaneous activation of all BDC subsets with a combination of R848 + poly(I:C) generates highly immunostimulatory DCs. These results support further investigation and clinical testing, as standalone or in conjunction with other immunotherapeutic strategies including adoptive T-cell transfer and checkpoint inhibition. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00262-021-03109-w.
format Online
Article
Text
id pubmed-8614222
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Springer Berlin Heidelberg
record_format MEDLINE/PubMed
spelling pubmed-86142222021-11-26 Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens Hänel, Gerulf Angerer, Caroline Petry, Katja Lichtenegger, Felix S. Subklewe, Marion Cancer Immunol Immunother Original Article Monocyte-derived Dendritic cells (DCs) have successfully been employed to induce immune responses against tumor-associated antigens in patients with various cancer entities. However, objective clinical responses have only been achieved in a minority of patients. Additionally, generation of GMP-compliant DCs requires time- and labor-intensive cell differentiation. In contrast, Blood DCs (BDCs) require only minimal ex vivo handling, as differentiation occurs in vivo resulting in potentially better functional capacities and survival. We aimed to identify a protocol for optimal in vitro activation of BDCs including the three subsets pDCs, cDC1s, and cDC2s. We evaluated several TLR ligand combinations and demonstrated that polyinosinic:polycytidylic acid [poly(I:C)] and R848, ligands for TLR3 and TLR7/8, respectively, constituted the optimal combination for inducing a positive co-stimulatory profile in all BDC subsets. In addition, TLR3 and TLR7/8 activation led to high secretion of IFN-α and IL-12p70. Simultaneous as opposed to separate tailored activation of pDCs and cDCs increased immunostimulatory capacities, suggesting that BDC subsets engage in synergistic cross-talk during activation. Stimulation of BDCs with this protocol resulted in enhanced migration, high NK-cell activation, and potent antigen-specific T-cell induction. We conclude that simultaneous activation of all BDC subsets with a combination of R848 + poly(I:C) generates highly immunostimulatory DCs. These results support further investigation and clinical testing, as standalone or in conjunction with other immunotherapeutic strategies including adoptive T-cell transfer and checkpoint inhibition. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00262-021-03109-w. Springer Berlin Heidelberg 2021-11-25 2022 /pmc/articles/PMC8614222/ /pubmed/34821951 http://dx.doi.org/10.1007/s00262-021-03109-w Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Original Article
Hänel, Gerulf
Angerer, Caroline
Petry, Katja
Lichtenegger, Felix S.
Subklewe, Marion
Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens
title Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens
title_full Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens
title_fullStr Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens
title_full_unstemmed Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens
title_short Blood DCs activated with R848 and poly(I:C) induce antigen-specific immune responses against viral and tumor-associated antigens
title_sort blood dcs activated with r848 and poly(i:c) induce antigen-specific immune responses against viral and tumor-associated antigens
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8614222/
https://www.ncbi.nlm.nih.gov/pubmed/34821951
http://dx.doi.org/10.1007/s00262-021-03109-w
work_keys_str_mv AT hanelgerulf blooddcsactivatedwithr848andpolyicinduceantigenspecificimmuneresponsesagainstviralandtumorassociatedantigens
AT angerercaroline blooddcsactivatedwithr848andpolyicinduceantigenspecificimmuneresponsesagainstviralandtumorassociatedantigens
AT petrykatja blooddcsactivatedwithr848andpolyicinduceantigenspecificimmuneresponsesagainstviralandtumorassociatedantigens
AT lichteneggerfelixs blooddcsactivatedwithr848andpolyicinduceantigenspecificimmuneresponsesagainstviralandtumorassociatedantigens
AT subklewemarion blooddcsactivatedwithr848andpolyicinduceantigenspecificimmuneresponsesagainstviralandtumorassociatedantigens