Cargando…

Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models

Autologous patient-derived dendritic cells (DCs) modified ex vivo to present tumor-associated antigens (TAAs) are frequently used as cancer vaccines. However, apart from the stringent logistics in producing DCs on a patient basis, accumulating evidence indicate that ex vivo engineered DCs are poor i...

Descripción completa

Detalles Bibliográficos
Autores principales: Fotaki, Grammatiki, Jin, Chuan, Kerzeli, Iliana Kyriaki, Ramachandran, Mohanraj, Martikainen, Minttu-Maria, Karlsson-Parra, Alex, Yu, Di, Essand, Magnus
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Taylor & Francis 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5790347/
https://www.ncbi.nlm.nih.gov/pubmed/29399398
http://dx.doi.org/10.1080/2162402X.2017.1397250
_version_ 1783296435746242560
author Fotaki, Grammatiki
Jin, Chuan
Kerzeli, Iliana Kyriaki
Ramachandran, Mohanraj
Martikainen, Minttu-Maria
Karlsson-Parra, Alex
Yu, Di
Essand, Magnus
author_facet Fotaki, Grammatiki
Jin, Chuan
Kerzeli, Iliana Kyriaki
Ramachandran, Mohanraj
Martikainen, Minttu-Maria
Karlsson-Parra, Alex
Yu, Di
Essand, Magnus
author_sort Fotaki, Grammatiki
collection PubMed
description Autologous patient-derived dendritic cells (DCs) modified ex vivo to present tumor-associated antigens (TAAs) are frequently used as cancer vaccines. However, apart from the stringent logistics in producing DCs on a patient basis, accumulating evidence indicate that ex vivo engineered DCs are poor in migration and in fact do not directly present TAA epitopes to naïve T cells in vivo. Instead, it is proposed that bystander host DCs take up material from vaccine-DCs, migrate and subsequently initiate antitumor T-cell responses. We used mouse models to examine the possibility of using pro-inflammatory allogeneic DCs (alloDCs) to activate host DCs and enable them to promote antigen-specific T-cell immunity. We found that alloDCs were able to initiate host DC activation and migration to draining lymph node leading to T-cell activation. The pro-inflammatory milieu created by alloDCs also led to recruitment of NK cells and neutrophils at the site of injection. Vaccination with alloDCs combined with Ad5M(gp100), an infection-enhanced adenovirus encoding the human melanoma-associated antigen gp100 resulted in generation of CD8(+) T cells with a T-cell receptor (TCR) specific for the gp100(25-33) epitope (gp100-TCR(+)). Ad5M(gp100)-alloDC vaccination in combination with transfer of gp100-specific pmel-1 T cells resulted in prolonged survival of B16-F10 melanoma-bearing mice and altered the composition of the tumor microenvironment (TME). We hereby propose that alloDCs together with TAA- or neoepitope-encoding Ad5M can become an “off-the-shelf” cancer vaccine, which can reverse the TME-induced immunosuppression and induce host cellular anti-tumor immune responses in patients without the need of a time-consuming preparation step of autologous DCs.
format Online
Article
Text
id pubmed-5790347
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Taylor & Francis
record_format MEDLINE/PubMed
spelling pubmed-57903472018-02-02 Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models Fotaki, Grammatiki Jin, Chuan Kerzeli, Iliana Kyriaki Ramachandran, Mohanraj Martikainen, Minttu-Maria Karlsson-Parra, Alex Yu, Di Essand, Magnus Oncoimmunology Original Research Autologous patient-derived dendritic cells (DCs) modified ex vivo to present tumor-associated antigens (TAAs) are frequently used as cancer vaccines. However, apart from the stringent logistics in producing DCs on a patient basis, accumulating evidence indicate that ex vivo engineered DCs are poor in migration and in fact do not directly present TAA epitopes to naïve T cells in vivo. Instead, it is proposed that bystander host DCs take up material from vaccine-DCs, migrate and subsequently initiate antitumor T-cell responses. We used mouse models to examine the possibility of using pro-inflammatory allogeneic DCs (alloDCs) to activate host DCs and enable them to promote antigen-specific T-cell immunity. We found that alloDCs were able to initiate host DC activation and migration to draining lymph node leading to T-cell activation. The pro-inflammatory milieu created by alloDCs also led to recruitment of NK cells and neutrophils at the site of injection. Vaccination with alloDCs combined with Ad5M(gp100), an infection-enhanced adenovirus encoding the human melanoma-associated antigen gp100 resulted in generation of CD8(+) T cells with a T-cell receptor (TCR) specific for the gp100(25-33) epitope (gp100-TCR(+)). Ad5M(gp100)-alloDC vaccination in combination with transfer of gp100-specific pmel-1 T cells resulted in prolonged survival of B16-F10 melanoma-bearing mice and altered the composition of the tumor microenvironment (TME). We hereby propose that alloDCs together with TAA- or neoepitope-encoding Ad5M can become an “off-the-shelf” cancer vaccine, which can reverse the TME-induced immunosuppression and induce host cellular anti-tumor immune responses in patients without the need of a time-consuming preparation step of autologous DCs. Taylor & Francis 2017-12-26 /pmc/articles/PMC5790347/ /pubmed/29399398 http://dx.doi.org/10.1080/2162402X.2017.1397250 Text en © 2018 The Author(s). Published with license by Taylor & Francis Group, LLC http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
spellingShingle Original Research
Fotaki, Grammatiki
Jin, Chuan
Kerzeli, Iliana Kyriaki
Ramachandran, Mohanraj
Martikainen, Minttu-Maria
Karlsson-Parra, Alex
Yu, Di
Essand, Magnus
Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models
title Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models
title_full Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models
title_fullStr Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models
title_full_unstemmed Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models
title_short Cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic DCs leads to sustained antigen-specific immune responses in three melanoma models
title_sort cancer vaccine based on a combination of an infection-enhanced adenoviral vector and pro-inflammatory allogeneic dcs leads to sustained antigen-specific immune responses in three melanoma models
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5790347/
https://www.ncbi.nlm.nih.gov/pubmed/29399398
http://dx.doi.org/10.1080/2162402X.2017.1397250
work_keys_str_mv AT fotakigrammatiki cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels
AT jinchuan cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels
AT kerzeliilianakyriaki cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels
AT ramachandranmohanraj cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels
AT martikainenminttumaria cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels
AT karlssonparraalex cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels
AT yudi cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels
AT essandmagnus cancervaccinebasedonacombinationofaninfectionenhancedadenoviralvectorandproinflammatoryallogeneicdcsleadstosustainedantigenspecificimmuneresponsesinthreemelanomamodels