Cargando…

Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor

The tumor suppressor p53 responds to genotoxic and oncogenic stresses by inducing cell cycle arrest and apoptosis. Recent studies suggest that p53 also participates in the regulation of cellular immune responses. Here, we have investigated the potential of p53 gene therapy to augment immune checkpoi...

Descripción completa

Detalles Bibliográficos
Autores principales: Kim, Sang-Soo, Harford, Joe B., Moghe, Manish, Rait, Antonina, Chang, Esther H.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Taylor & Francis 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6169574/
https://www.ncbi.nlm.nih.gov/pubmed/30288347
http://dx.doi.org/10.1080/2162402X.2018.1484982
_version_ 1783360537830096896
author Kim, Sang-Soo
Harford, Joe B.
Moghe, Manish
Rait, Antonina
Chang, Esther H.
author_facet Kim, Sang-Soo
Harford, Joe B.
Moghe, Manish
Rait, Antonina
Chang, Esther H.
author_sort Kim, Sang-Soo
collection PubMed
description The tumor suppressor p53 responds to genotoxic and oncogenic stresses by inducing cell cycle arrest and apoptosis. Recent studies suggest that p53 also participates in the regulation of cellular immune responses. Here, we have investigated the potential of p53 gene therapy to augment immune checkpoint inhibition by combining an anti-programmed cell death protein 1 (PD1) antibody with SGT-53, our investigational nanomedicine carrying a plasmid encoding human wild-type p53. In three syngeneic mouse tumor models examined including a breast cancer, a non-small cell lung carcinoma, and a glioblastoma, SGT-53 sensitized otherwise refractory tumors to anti-PD1 antibody. The involvement of p53 in enhancing anti-PD1 immunotherapy appears to be multifaceted, since SGT-53 treatment increased tumor immunogenicity, enhanced both innate and adaptive immune responses, and reduced tumor-induced immunosuppression in a 4T1 breast tumor model. In addition, SGT-53 alleviates a fatal xenogeneic hypersensitivity associated with the anti-PD1 antibody in this model. Our data suggest that restoring p53 function by SGT-53 is able to boost anti-tumor immunity to augment anti-PD1 therapy by sensitizing tumors otherwise insensitive to anti-PD1 immunotherapy while reducing immune-related adverse events.
format Online
Article
Text
id pubmed-6169574
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Taylor & Francis
record_format MEDLINE/PubMed
spelling pubmed-61695742018-10-04 Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor Kim, Sang-Soo Harford, Joe B. Moghe, Manish Rait, Antonina Chang, Esther H. Oncoimmunology Original Research The tumor suppressor p53 responds to genotoxic and oncogenic stresses by inducing cell cycle arrest and apoptosis. Recent studies suggest that p53 also participates in the regulation of cellular immune responses. Here, we have investigated the potential of p53 gene therapy to augment immune checkpoint inhibition by combining an anti-programmed cell death protein 1 (PD1) antibody with SGT-53, our investigational nanomedicine carrying a plasmid encoding human wild-type p53. In three syngeneic mouse tumor models examined including a breast cancer, a non-small cell lung carcinoma, and a glioblastoma, SGT-53 sensitized otherwise refractory tumors to anti-PD1 antibody. The involvement of p53 in enhancing anti-PD1 immunotherapy appears to be multifaceted, since SGT-53 treatment increased tumor immunogenicity, enhanced both innate and adaptive immune responses, and reduced tumor-induced immunosuppression in a 4T1 breast tumor model. In addition, SGT-53 alleviates a fatal xenogeneic hypersensitivity associated with the anti-PD1 antibody in this model. Our data suggest that restoring p53 function by SGT-53 is able to boost anti-tumor immunity to augment anti-PD1 therapy by sensitizing tumors otherwise insensitive to anti-PD1 immunotherapy while reducing immune-related adverse events. Taylor & Francis 2018-08-01 /pmc/articles/PMC6169574/ /pubmed/30288347 http://dx.doi.org/10.1080/2162402X.2018.1484982 Text en © 2018 The Author(s). Published by Taylor & Francis. 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
Kim, Sang-Soo
Harford, Joe B.
Moghe, Manish
Rait, Antonina
Chang, Esther H.
Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor
title Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor
title_full Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor
title_fullStr Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor
title_full_unstemmed Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor
title_short Combination with SGT-53 overcomes tumor resistance to a checkpoint inhibitor
title_sort combination with sgt-53 overcomes tumor resistance to a checkpoint inhibitor
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6169574/
https://www.ncbi.nlm.nih.gov/pubmed/30288347
http://dx.doi.org/10.1080/2162402X.2018.1484982
work_keys_str_mv AT kimsangsoo combinationwithsgt53overcomestumorresistancetoacheckpointinhibitor
AT harfordjoeb combinationwithsgt53overcomestumorresistancetoacheckpointinhibitor
AT moghemanish combinationwithsgt53overcomestumorresistancetoacheckpointinhibitor
AT raitantonina combinationwithsgt53overcomestumorresistancetoacheckpointinhibitor
AT changestherh combinationwithsgt53overcomestumorresistancetoacheckpointinhibitor