Cargando…

Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation

Residual lesions and undetectable metastasis after insufficient radiofrequency ablation (iRFA) are associated with earlier new metastases and poor survival in cancer patients, for induced aggressive tumor phenotype and immunosuppression. Programmed cell death protein 1(PD-1) blockade has been report...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Tongyi, Wu, Wenxin, Wu, Jiancong, Tan, Yang, Zhang, Minru, Long, Haiyi, Guo, Huanling, Zhang, Xiaoer, Zhou, Wenwen, Zhang, Qi, Xie, Xiaoyan, Xu, Ming, Zhang, Chunyang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10083362/
https://www.ncbi.nlm.nih.gov/pubmed/37051250
http://dx.doi.org/10.3389/fimmu.2023.1124152
_version_ 1785021493522989056
author Huang, Tongyi
Wu, Wenxin
Wu, Jiancong
Tan, Yang
Zhang, Minru
Long, Haiyi
Guo, Huanling
Zhang, Xiaoer
Zhou, Wenwen
Zhang, Qi
Xie, Xiaoyan
Xu, Ming
Zhang, Chunyang
author_facet Huang, Tongyi
Wu, Wenxin
Wu, Jiancong
Tan, Yang
Zhang, Minru
Long, Haiyi
Guo, Huanling
Zhang, Xiaoer
Zhou, Wenwen
Zhang, Qi
Xie, Xiaoyan
Xu, Ming
Zhang, Chunyang
author_sort Huang, Tongyi
collection PubMed
description Residual lesions and undetectable metastasis after insufficient radiofrequency ablation (iRFA) are associated with earlier new metastases and poor survival in cancer patients, for induced aggressive tumor phenotype and immunosuppression. Programmed cell death protein 1(PD-1) blockade has been reported to enhance the radiofrequency ablation-elicited antitumor immunity, but its ability to eliminate incompletely ablated residual lesions has been questioned. Here, we report a combined treatment modality post iRFA based on integrating an oxygen self-enriching nanodrug PFH-Ce6 liposome@O(2) nanodroplets (PCL@O(2))-augmented noninvasive sonodynamic therapy (SDT) with PD-1 blockade. PCL@O(2) containing Ce6 as the sonosensitizer and PFH as O(2) reservoir, was synthesized as an augmented SDT nanoplatform and showed increased ROS generation to raise effective apoptosis of tumor cells, which also exposed more calreticulin to induce stronger immunogenic cell death (ICD). Combining with PD-1 blockade post iRFA, this optimized SDT induced a better anti-tumor response in MC38 tumor bearing mouse model, which not only arrested residual primary tumor progression, but also inhibited the growth of distant tumor, therefore prolonging the survival. Profiling of immune populations within the tumor draining lymph nodes and tumors further revealed that combination therapy effectively induced ICD, and promoted the maturation of dendritic cells, tumor infiltration of T cells, as well as activation of cytotoxic T lymphocytes. While iRFA alone could result in an increase of regulatory T cells (Tregs) in the residual tumors, SDT plus PD-1 blockade post iRFA reduced the number of Tregs in both primary and distant tumors. Moreover, the combined treatment could significantly initiate long-term immune memory, manifesting as elevated levels of CD8(+) and CD4(+) central memory cells. Therefore, this study establishes the preclinical proof of concept to apply oxygen self-enriching SDT to augment cancer immunotherapy after iRFA.
format Online
Article
Text
id pubmed-10083362
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-100833622023-04-11 Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation Huang, Tongyi Wu, Wenxin Wu, Jiancong Tan, Yang Zhang, Minru Long, Haiyi Guo, Huanling Zhang, Xiaoer Zhou, Wenwen Zhang, Qi Xie, Xiaoyan Xu, Ming Zhang, Chunyang Front Immunol Immunology Residual lesions and undetectable metastasis after insufficient radiofrequency ablation (iRFA) are associated with earlier new metastases and poor survival in cancer patients, for induced aggressive tumor phenotype and immunosuppression. Programmed cell death protein 1(PD-1) blockade has been reported to enhance the radiofrequency ablation-elicited antitumor immunity, but its ability to eliminate incompletely ablated residual lesions has been questioned. Here, we report a combined treatment modality post iRFA based on integrating an oxygen self-enriching nanodrug PFH-Ce6 liposome@O(2) nanodroplets (PCL@O(2))-augmented noninvasive sonodynamic therapy (SDT) with PD-1 blockade. PCL@O(2) containing Ce6 as the sonosensitizer and PFH as O(2) reservoir, was synthesized as an augmented SDT nanoplatform and showed increased ROS generation to raise effective apoptosis of tumor cells, which also exposed more calreticulin to induce stronger immunogenic cell death (ICD). Combining with PD-1 blockade post iRFA, this optimized SDT induced a better anti-tumor response in MC38 tumor bearing mouse model, which not only arrested residual primary tumor progression, but also inhibited the growth of distant tumor, therefore prolonging the survival. Profiling of immune populations within the tumor draining lymph nodes and tumors further revealed that combination therapy effectively induced ICD, and promoted the maturation of dendritic cells, tumor infiltration of T cells, as well as activation of cytotoxic T lymphocytes. While iRFA alone could result in an increase of regulatory T cells (Tregs) in the residual tumors, SDT plus PD-1 blockade post iRFA reduced the number of Tregs in both primary and distant tumors. Moreover, the combined treatment could significantly initiate long-term immune memory, manifesting as elevated levels of CD8(+) and CD4(+) central memory cells. Therefore, this study establishes the preclinical proof of concept to apply oxygen self-enriching SDT to augment cancer immunotherapy after iRFA. Frontiers Media S.A. 2023-03-27 /pmc/articles/PMC10083362/ /pubmed/37051250 http://dx.doi.org/10.3389/fimmu.2023.1124152 Text en Copyright © 2023 Huang, Wu, Wu, Tan, Zhang, Long, Guo, Zhang, Zhou, Zhang, Xie, Xu and Zhang 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
Huang, Tongyi
Wu, Wenxin
Wu, Jiancong
Tan, Yang
Zhang, Minru
Long, Haiyi
Guo, Huanling
Zhang, Xiaoer
Zhou, Wenwen
Zhang, Qi
Xie, Xiaoyan
Xu, Ming
Zhang, Chunyang
Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation
title Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation
title_full Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation
title_fullStr Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation
title_full_unstemmed Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation
title_short Perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation
title_sort perfluorocarbon nanodrug induced oxygen self-enriching sonodynamic therapy improves cancer immunotherapy after insufficient radiofrequency ablation
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10083362/
https://www.ncbi.nlm.nih.gov/pubmed/37051250
http://dx.doi.org/10.3389/fimmu.2023.1124152
work_keys_str_mv AT huangtongyi perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT wuwenxin perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT wujiancong perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT tanyang perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT zhangminru perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT longhaiyi perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT guohuanling perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT zhangxiaoer perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT zhouwenwen perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT zhangqi perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT xiexiaoyan perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT xuming perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation
AT zhangchunyang perfluorocarbonnanodruginducedoxygenselfenrichingsonodynamictherapyimprovescancerimmunotherapyafterinsufficientradiofrequencyablation