Cargando…

PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis

Metastasis is the main cause of death in individuals with cancer. Immune checkpoint blockade (ICB) can potentially reverse CD8(+) cytotoxic T lymphocytes (CTLs) dysfunction, leading to significant remission in multiple cancers. However, the mechanism underlying the development of CTL exhaustion duri...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Ji, Song, Yang, Miao, Feng, Chen, Gang, Zhu, Yongjun, Wu, Ning, Pang, Liewen, Chen, Zhiming, Chen, Xiaofeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8409314/
https://www.ncbi.nlm.nih.gov/pubmed/34152672
http://dx.doi.org/10.1111/cas.15033
_version_ 1783746974460149760
author Chen, Ji
Song, Yang
Miao, Feng
Chen, Gang
Zhu, Yongjun
Wu, Ning
Pang, Liewen
Chen, Zhiming
Chen, Xiaofeng
author_facet Chen, Ji
Song, Yang
Miao, Feng
Chen, Gang
Zhu, Yongjun
Wu, Ning
Pang, Liewen
Chen, Zhiming
Chen, Xiaofeng
author_sort Chen, Ji
collection PubMed
description Metastasis is the main cause of death in individuals with cancer. Immune checkpoint blockade (ICB) can potentially reverse CD8(+) cytotoxic T lymphocytes (CTLs) dysfunction, leading to significant remission in multiple cancers. However, the mechanism underlying the development of CTL exhaustion during metastatic progression remains unclear. Here, we established an experimental pulmonary metastasis model with melanoma cells and discovered a critical role for melanoma‐released exosomes in metastasis. Using genetic knockdown of nSMase2 and Rab27a, 2 key enzymes for exosome secretion, we showed that high levels of effector‐like tumor‐specific CD8(+) T cells with transitory exhaustion, instead of terminal exhaustion, were observed in mice without exosomes; these cells showed limited inhibitory receptors and strong proliferation and cytotoxicity. Mechanistically, the immunosuppression of exosomes depends on exogenous PD‐L1, which can be largely rescued by pretreatment with antibody blockade. Notably, we also found that exosomal PD‐L1 acts as a promising predictive biomarker for ICB therapies during metastasis. Together, our findings suggest that exosomal PD‐L1 may be a potential immunotherapy target, suggesting a new curative therapy for tumor metastasis.
format Online
Article
Text
id pubmed-8409314
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-84093142021-09-03 PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis Chen, Ji Song, Yang Miao, Feng Chen, Gang Zhu, Yongjun Wu, Ning Pang, Liewen Chen, Zhiming Chen, Xiaofeng Cancer Sci Original Articles Metastasis is the main cause of death in individuals with cancer. Immune checkpoint blockade (ICB) can potentially reverse CD8(+) cytotoxic T lymphocytes (CTLs) dysfunction, leading to significant remission in multiple cancers. However, the mechanism underlying the development of CTL exhaustion during metastatic progression remains unclear. Here, we established an experimental pulmonary metastasis model with melanoma cells and discovered a critical role for melanoma‐released exosomes in metastasis. Using genetic knockdown of nSMase2 and Rab27a, 2 key enzymes for exosome secretion, we showed that high levels of effector‐like tumor‐specific CD8(+) T cells with transitory exhaustion, instead of terminal exhaustion, were observed in mice without exosomes; these cells showed limited inhibitory receptors and strong proliferation and cytotoxicity. Mechanistically, the immunosuppression of exosomes depends on exogenous PD‐L1, which can be largely rescued by pretreatment with antibody blockade. Notably, we also found that exosomal PD‐L1 acts as a promising predictive biomarker for ICB therapies during metastasis. Together, our findings suggest that exosomal PD‐L1 may be a potential immunotherapy target, suggesting a new curative therapy for tumor metastasis. John Wiley and Sons Inc. 2021-07-29 2021-09 /pmc/articles/PMC8409314/ /pubmed/34152672 http://dx.doi.org/10.1111/cas.15033 Text en © 2021 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ (https://creativecommons.org/licenses/by-nc-nd/4.0/) License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.
spellingShingle Original Articles
Chen, Ji
Song, Yang
Miao, Feng
Chen, Gang
Zhu, Yongjun
Wu, Ning
Pang, Liewen
Chen, Zhiming
Chen, Xiaofeng
PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis
title PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis
title_full PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis
title_fullStr PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis
title_full_unstemmed PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis
title_short PDL1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific CD8(+) T cell exhaustion during metastasis
title_sort pdl1‐positive exosomes suppress antitumor immunity by inducing tumor‐specific cd8(+) t cell exhaustion during metastasis
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8409314/
https://www.ncbi.nlm.nih.gov/pubmed/34152672
http://dx.doi.org/10.1111/cas.15033
work_keys_str_mv AT chenji pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT songyang pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT miaofeng pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT chengang pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT zhuyongjun pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT wuning pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT pangliewen pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT chenzhiming pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis
AT chenxiaofeng pdl1positiveexosomessuppressantitumorimmunitybyinducingtumorspecificcd8tcellexhaustionduringmetastasis