Cargando…

Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine

INTRODUCTION: Tumor-specific mutations generate neoepitopes unique to the cancer that can be recognized by the immune system, making them appealing targets for therapeutic cancer vaccines. Since the vast majority of tumor mutations are patient-specific, it is crucial for cancer vaccine designs to be...

Descripción completa

Detalles Bibliográficos
Autores principales: Barrio-Calvo, Marina, Kofoed, Søren Vester, Holste, Sofie Cens, Sørensen, Anders Bundgård, Viborg, Nadia, Kringelum, Jens Vindahl, Kleine-Kohlbrecher, Daniela, Steenmans, Christian Skjødt, Thygesen, Christian Bahne, Rønø, Birgitte, Friis, Stine
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10499626/
https://www.ncbi.nlm.nih.gov/pubmed/37720215
http://dx.doi.org/10.3389/fimmu.2023.1234912
_version_ 1785105750388899840
author Barrio-Calvo, Marina
Kofoed, Søren Vester
Holste, Sofie Cens
Sørensen, Anders Bundgård
Viborg, Nadia
Kringelum, Jens Vindahl
Kleine-Kohlbrecher, Daniela
Steenmans, Christian Skjødt
Thygesen, Christian Bahne
Rønø, Birgitte
Friis, Stine
author_facet Barrio-Calvo, Marina
Kofoed, Søren Vester
Holste, Sofie Cens
Sørensen, Anders Bundgård
Viborg, Nadia
Kringelum, Jens Vindahl
Kleine-Kohlbrecher, Daniela
Steenmans, Christian Skjødt
Thygesen, Christian Bahne
Rønø, Birgitte
Friis, Stine
author_sort Barrio-Calvo, Marina
collection PubMed
description INTRODUCTION: Tumor-specific mutations generate neoepitopes unique to the cancer that can be recognized by the immune system, making them appealing targets for therapeutic cancer vaccines. Since the vast majority of tumor mutations are patient-specific, it is crucial for cancer vaccine designs to be compatible with individualized treatment strategies. Plasmid DNA vaccines have substantiated the immunogenicity and tumor eradication capacity of cancer neoepitopes in preclinical models. Moreover, early clinical trials evaluating personalized neoepitope vaccines have indicated favorable safety profiles and demonstrated their ability to elicit specific immune responses toward the vaccine neoepitopes. METHODS: By fusing in silico predicted neoepitopes to molecules with affinity for receptors on the surface of APCs, such as chemokine (C-C motif) ligand 19 (CCL19), we designed an APC-targeting cancer vaccine and evaluated their ability to induce T-cell responses and anti-tumor efficacy in the BALB/c syngeneic preclinical tumor model. RESULTS: In this study, we demonstrate how the addition of an antigen-presenting cell (APC) binding molecule to DNA-encoded cancer neoepitopes improves neoepitope-specific T-cell responses and the anti-tumor efficacy of plasmid DNA vaccines. Dose-response evaluation and longitudinal analysis of neoepitope-specific T-cell responses indicate that combining APC-binding molecules with the delivery of personalized tumor antigens holds the potential to improve the clinical efficacy of therapeutic DNA cancer vaccines. DISCUSSION: Our findings indicate the potential of the APC-targeting strategy to enhance personalized DNA cancer vaccines while acknowledging the need for further research to investigate its molecular mechanism of action and to translate the preclinical results into effective treatments for cancer patients.
format Online
Article
Text
id pubmed-10499626
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-104996262023-09-15 Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine Barrio-Calvo, Marina Kofoed, Søren Vester Holste, Sofie Cens Sørensen, Anders Bundgård Viborg, Nadia Kringelum, Jens Vindahl Kleine-Kohlbrecher, Daniela Steenmans, Christian Skjødt Thygesen, Christian Bahne Rønø, Birgitte Friis, Stine Front Immunol Immunology INTRODUCTION: Tumor-specific mutations generate neoepitopes unique to the cancer that can be recognized by the immune system, making them appealing targets for therapeutic cancer vaccines. Since the vast majority of tumor mutations are patient-specific, it is crucial for cancer vaccine designs to be compatible with individualized treatment strategies. Plasmid DNA vaccines have substantiated the immunogenicity and tumor eradication capacity of cancer neoepitopes in preclinical models. Moreover, early clinical trials evaluating personalized neoepitope vaccines have indicated favorable safety profiles and demonstrated their ability to elicit specific immune responses toward the vaccine neoepitopes. METHODS: By fusing in silico predicted neoepitopes to molecules with affinity for receptors on the surface of APCs, such as chemokine (C-C motif) ligand 19 (CCL19), we designed an APC-targeting cancer vaccine and evaluated their ability to induce T-cell responses and anti-tumor efficacy in the BALB/c syngeneic preclinical tumor model. RESULTS: In this study, we demonstrate how the addition of an antigen-presenting cell (APC) binding molecule to DNA-encoded cancer neoepitopes improves neoepitope-specific T-cell responses and the anti-tumor efficacy of plasmid DNA vaccines. Dose-response evaluation and longitudinal analysis of neoepitope-specific T-cell responses indicate that combining APC-binding molecules with the delivery of personalized tumor antigens holds the potential to improve the clinical efficacy of therapeutic DNA cancer vaccines. DISCUSSION: Our findings indicate the potential of the APC-targeting strategy to enhance personalized DNA cancer vaccines while acknowledging the need for further research to investigate its molecular mechanism of action and to translate the preclinical results into effective treatments for cancer patients. Frontiers Media S.A. 2023-08-29 /pmc/articles/PMC10499626/ /pubmed/37720215 http://dx.doi.org/10.3389/fimmu.2023.1234912 Text en Copyright © 2023 Barrio-Calvo, Kofoed, Holste, Sørensen, Viborg, Kringelum, Kleine-Kohlbrecher, Steenmans, Thygesen, Rønø and Friis 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 Immunology
Barrio-Calvo, Marina
Kofoed, Søren Vester
Holste, Sofie Cens
Sørensen, Anders Bundgård
Viborg, Nadia
Kringelum, Jens Vindahl
Kleine-Kohlbrecher, Daniela
Steenmans, Christian Skjødt
Thygesen, Christian Bahne
Rønø, Birgitte
Friis, Stine
Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine
title Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine
title_full Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine
title_fullStr Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine
title_full_unstemmed Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine
title_short Targeting neoantigens to APC-surface molecules improves the immunogenicity and anti-tumor efficacy of a DNA cancer vaccine
title_sort targeting neoantigens to apc-surface molecules improves the immunogenicity and anti-tumor efficacy of a dna cancer vaccine
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10499626/
https://www.ncbi.nlm.nih.gov/pubmed/37720215
http://dx.doi.org/10.3389/fimmu.2023.1234912
work_keys_str_mv AT barriocalvomarina targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT kofoedsørenvester targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT holstesofiecens targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT sørensenandersbundgard targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT viborgnadia targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT kringelumjensvindahl targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT kleinekohlbrecherdaniela targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT steenmanschristianskjødt targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT thygesenchristianbahne targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT rønøbirgitte targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine
AT friisstine targetingneoantigenstoapcsurfacemoleculesimprovestheimmunogenicityandantitumorefficacyofadnacancervaccine