Cargando…

A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models

Lung cancer is among the most common and lethal cancers and warrants novel therapeutic approaches to improving patient outcomes. Although immune checkpoint inhibitors (ICIs) have demonstrated substantial clinical benefits, most patients remain unresponsive to currently approved ICIs or develop resis...

Descripción completa

Detalles Bibliográficos
Autores principales: Kim, Sang-Soo, Harford, Joe B., Moghe, Manish, Doherty, Caroline, Chang, Esther H.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9654894/
https://www.ncbi.nlm.nih.gov/pubmed/36359830
http://dx.doi.org/10.3390/cells11213434
_version_ 1784829048962154496
author Kim, Sang-Soo
Harford, Joe B.
Moghe, Manish
Doherty, Caroline
Chang, Esther H.
author_facet Kim, Sang-Soo
Harford, Joe B.
Moghe, Manish
Doherty, Caroline
Chang, Esther H.
author_sort Kim, Sang-Soo
collection PubMed
description Lung cancer is among the most common and lethal cancers and warrants novel therapeutic approaches to improving patient outcomes. Although immune checkpoint inhibitors (ICIs) have demonstrated substantial clinical benefits, most patients remain unresponsive to currently approved ICIs or develop resistance after initial response. Many ongoing clinical studies are investigating combination therapies to address the limited efficacy of ICIs. Here, we have assessed whether p53 gene therapy via a tumor-targeting nanomedicine (termed SGT-53) can augment anti-programmed cell death-1 (PD-1) immunotherapy to expand its use in non-responding patients. Using syngeneic mouse models of lung cancers that are resistant to anti-PD-1, we demonstrate that restoration of normal p53 function potentiates anti-PD-1 to inhibit tumor growth and prolong survival of tumor-bearing animals. Our data indicate that SGT-53 can restore effective immune responses against lung cancer cells by reducing immuno-suppressive cells (M2 macrophages and regulatory T cells) and by downregulating immunosuppressive molecules (e.g., galectin-1, a negative regulator of T cell activation and survival) while increasing activity of cytotoxic T cells. These results suggest that combining SGT-53 with anti-PD-1 immunotherapy could increase the fraction of lung cancer patients that responds to anti-PD-1 therapy and support evaluation of this combination particularly in patients with ICI-resistant lung cancers.
format Online
Article
Text
id pubmed-9654894
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-96548942022-11-15 A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models Kim, Sang-Soo Harford, Joe B. Moghe, Manish Doherty, Caroline Chang, Esther H. Cells Article Lung cancer is among the most common and lethal cancers and warrants novel therapeutic approaches to improving patient outcomes. Although immune checkpoint inhibitors (ICIs) have demonstrated substantial clinical benefits, most patients remain unresponsive to currently approved ICIs or develop resistance after initial response. Many ongoing clinical studies are investigating combination therapies to address the limited efficacy of ICIs. Here, we have assessed whether p53 gene therapy via a tumor-targeting nanomedicine (termed SGT-53) can augment anti-programmed cell death-1 (PD-1) immunotherapy to expand its use in non-responding patients. Using syngeneic mouse models of lung cancers that are resistant to anti-PD-1, we demonstrate that restoration of normal p53 function potentiates anti-PD-1 to inhibit tumor growth and prolong survival of tumor-bearing animals. Our data indicate that SGT-53 can restore effective immune responses against lung cancer cells by reducing immuno-suppressive cells (M2 macrophages and regulatory T cells) and by downregulating immunosuppressive molecules (e.g., galectin-1, a negative regulator of T cell activation and survival) while increasing activity of cytotoxic T cells. These results suggest that combining SGT-53 with anti-PD-1 immunotherapy could increase the fraction of lung cancer patients that responds to anti-PD-1 therapy and support evaluation of this combination particularly in patients with ICI-resistant lung cancers. MDPI 2022-10-31 /pmc/articles/PMC9654894/ /pubmed/36359830 http://dx.doi.org/10.3390/cells11213434 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Kim, Sang-Soo
Harford, Joe B.
Moghe, Manish
Doherty, Caroline
Chang, Esther H.
A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models
title A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models
title_full A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models
title_fullStr A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models
title_full_unstemmed A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models
title_short A Novel P53 Nanomedicine Reduces Immunosuppression and Augments Anti-PD-1 Therapy for Non-Small Cell Lung Cancer in Syngeneic Mouse Models
title_sort novel p53 nanomedicine reduces immunosuppression and augments anti-pd-1 therapy for non-small cell lung cancer in syngeneic mouse models
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9654894/
https://www.ncbi.nlm.nih.gov/pubmed/36359830
http://dx.doi.org/10.3390/cells11213434
work_keys_str_mv AT kimsangsoo anovelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT harfordjoeb anovelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT moghemanish anovelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT dohertycaroline anovelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT changestherh anovelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT kimsangsoo novelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT harfordjoeb novelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT moghemanish novelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT dohertycaroline novelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels
AT changestherh novelp53nanomedicinereducesimmunosuppressionandaugmentsantipd1therapyfornonsmallcelllungcancerinsyngeneicmousemodels