Cargando…

Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor

Cancer immunotherapy including adoptive T cell therapy (ACT) is widely used in the clinic and is highly beneficial for patients with hematological malignancies; however, it remains a challenge to develop effective immunotherapy strategies for the treatment of solid cancers, due to the inefficiency o...

Descripción completa

Detalles Bibliográficos
Autores principales: Jiang, Qingzhi, Zhang, Chi, Wang, Huilan, Peng, Tao, Zhang, Li, Wang, Yang, Han, Weidong, Shi, Chunmeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6861368/
https://www.ncbi.nlm.nih.gov/pubmed/31781498
http://dx.doi.org/10.3389/fonc.2019.01196
_version_ 1783471340700827648
author Jiang, Qingzhi
Zhang, Chi
Wang, Huilan
Peng, Tao
Zhang, Li
Wang, Yang
Han, Weidong
Shi, Chunmeng
author_facet Jiang, Qingzhi
Zhang, Chi
Wang, Huilan
Peng, Tao
Zhang, Li
Wang, Yang
Han, Weidong
Shi, Chunmeng
author_sort Jiang, Qingzhi
collection PubMed
description Cancer immunotherapy including adoptive T cell therapy (ACT) is widely used in the clinic and is highly beneficial for patients with hematological malignancies; however, it remains a challenge to develop effective immunotherapy strategies for the treatment of solid cancers, due to the inefficiency of the immune response and the immunosuppressive tumor microenvironment (TME). Immunogenic cell death (ICD) converts dying cancer cells into a therapeutic vaccine and stimulate a systemic antigen-specific antitumor immune response, which can effectively subvert the immunosuppressive TME and enhance the efficiency of immune responses, relative to conventional immunotherapeutic regimens. However, the application of traditional inducers of ICD in anti-cancer immunotherapy has been limited because of low levels of ICD induction and a lack of tumor-targeting accumulation. Mitochondria are important for tumor-targeting strategies and have emerged as organelles with key roles in the immune system. We hypothesized that the alteration of mitochondria in cancer cells could be an important target for the development of an efficient ICD inducer for use in cancer immunotherapy. Here, we report the evaluation of a mitochondria-targeted small molecule, IR-780, that acts as an ICD inducer and exhibits exceptional antineoplastic activity. IR-780 specifically accumulated in tumor cells to elicit ICD in vitro and in vivo, effectively suppressed tumor growth and lung metastasis, and enhanced adoptive T-cell therapy effects against solid tumors in mouse models. These anticancer effects were linked to dendritic cell maturation and synergistic effector T cell priming and infiltration into tumors. The underlying mechanism involves the direct targeting of the mitochondria by IR-780, to destroy cancer cells, including drug-resistant cancer cells, leading to the full exposure of tumor-associated antigens (TAAs), thereby enhancing antigen-specific antitumor immune responses. These features of IR-780 suggest that it has the advantage of leading to complete TAA exposure and the stimulation of efficient antitumor immune responses in the TME. IR-780 has potential for use as a preparative ICD inducer, in combination with conventional immunostimulatory regimens for cancer immunotherapy, particularly in the context of solid tumor treatment.
format Online
Article
Text
id pubmed-6861368
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-68613682019-11-28 Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor Jiang, Qingzhi Zhang, Chi Wang, Huilan Peng, Tao Zhang, Li Wang, Yang Han, Weidong Shi, Chunmeng Front Oncol Oncology Cancer immunotherapy including adoptive T cell therapy (ACT) is widely used in the clinic and is highly beneficial for patients with hematological malignancies; however, it remains a challenge to develop effective immunotherapy strategies for the treatment of solid cancers, due to the inefficiency of the immune response and the immunosuppressive tumor microenvironment (TME). Immunogenic cell death (ICD) converts dying cancer cells into a therapeutic vaccine and stimulate a systemic antigen-specific antitumor immune response, which can effectively subvert the immunosuppressive TME and enhance the efficiency of immune responses, relative to conventional immunotherapeutic regimens. However, the application of traditional inducers of ICD in anti-cancer immunotherapy has been limited because of low levels of ICD induction and a lack of tumor-targeting accumulation. Mitochondria are important for tumor-targeting strategies and have emerged as organelles with key roles in the immune system. We hypothesized that the alteration of mitochondria in cancer cells could be an important target for the development of an efficient ICD inducer for use in cancer immunotherapy. Here, we report the evaluation of a mitochondria-targeted small molecule, IR-780, that acts as an ICD inducer and exhibits exceptional antineoplastic activity. IR-780 specifically accumulated in tumor cells to elicit ICD in vitro and in vivo, effectively suppressed tumor growth and lung metastasis, and enhanced adoptive T-cell therapy effects against solid tumors in mouse models. These anticancer effects were linked to dendritic cell maturation and synergistic effector T cell priming and infiltration into tumors. The underlying mechanism involves the direct targeting of the mitochondria by IR-780, to destroy cancer cells, including drug-resistant cancer cells, leading to the full exposure of tumor-associated antigens (TAAs), thereby enhancing antigen-specific antitumor immune responses. These features of IR-780 suggest that it has the advantage of leading to complete TAA exposure and the stimulation of efficient antitumor immune responses in the TME. IR-780 has potential for use as a preparative ICD inducer, in combination with conventional immunostimulatory regimens for cancer immunotherapy, particularly in the context of solid tumor treatment. Frontiers Media S.A. 2019-11-12 /pmc/articles/PMC6861368/ /pubmed/31781498 http://dx.doi.org/10.3389/fonc.2019.01196 Text en Copyright © 2019 Jiang, Zhang, Wang, Peng, Zhang, Wang, Han and Shi. http://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 Oncology
Jiang, Qingzhi
Zhang, Chi
Wang, Huilan
Peng, Tao
Zhang, Li
Wang, Yang
Han, Weidong
Shi, Chunmeng
Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor
title Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor
title_full Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor
title_fullStr Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor
title_full_unstemmed Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor
title_short Mitochondria-Targeting Immunogenic Cell Death Inducer Improves the Adoptive T-Cell Therapy Against Solid Tumor
title_sort mitochondria-targeting immunogenic cell death inducer improves the adoptive t-cell therapy against solid tumor
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6861368/
https://www.ncbi.nlm.nih.gov/pubmed/31781498
http://dx.doi.org/10.3389/fonc.2019.01196
work_keys_str_mv AT jiangqingzhi mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor
AT zhangchi mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor
AT wanghuilan mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor
AT pengtao mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor
AT zhangli mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor
AT wangyang mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor
AT hanweidong mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor
AT shichunmeng mitochondriatargetingimmunogeniccelldeathinducerimprovestheadoptivetcelltherapyagainstsolidtumor