Cargando…

M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance

T-cell-based immune checkpoint blockade therapy (ICB) can be undermined by local immunosuppressive M2-like tumor-associated macrophages (TAMs). However, modulating macrophages has proved difficult as the molecular and functional features of M2-TAMs on tumor growth are still uncertain. Here we report...

Descripción completa

Detalles Bibliográficos
Autores principales: Zheng, Naisheng, Wang, Tingting, Luo, Qin, Liu, Yi, Yang, Junyao, Zhou, Yunlan, Xie, Guohua, Ma, Yanhui, Yuan, Xiangliang, Shen, Lisong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Taylor & Francis 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10184604/
https://www.ncbi.nlm.nih.gov/pubmed/37197441
http://dx.doi.org/10.1080/2162402X.2023.2210959
_version_ 1785042178942173184
author Zheng, Naisheng
Wang, Tingting
Luo, Qin
Liu, Yi
Yang, Junyao
Zhou, Yunlan
Xie, Guohua
Ma, Yanhui
Yuan, Xiangliang
Shen, Lisong
author_facet Zheng, Naisheng
Wang, Tingting
Luo, Qin
Liu, Yi
Yang, Junyao
Zhou, Yunlan
Xie, Guohua
Ma, Yanhui
Yuan, Xiangliang
Shen, Lisong
author_sort Zheng, Naisheng
collection PubMed
description T-cell-based immune checkpoint blockade therapy (ICB) can be undermined by local immunosuppressive M2-like tumor-associated macrophages (TAMs). However, modulating macrophages has proved difficult as the molecular and functional features of M2-TAMs on tumor growth are still uncertain. Here we reported that immunosuppressive M2 macrophages render cancer cells resistant to CD8(+) T-cell-dependent tumor-killing refractory ICB efficacy by secreting exosomes. Proteomics and functional studies revealed that M2 macrophage-derived exosome (M2-exo) transmitted apolipoprotein E (ApoE) to cancer cells conferring ICB resistance by downregulated MHC-I expression curbing tumor intrinsic immunogenicity. Mechanistically, M2 exosomal ApoE diminished the tumor-intrinsic ATPase activity of binding immunoglobulin protein (BiP) to decrease tumor MHC-I expression. Sensitizing ICB efficacy can be achieved by the administration of ApoE ligand, EZ-482, enhancing ATPase activity of BiP to boost tumor-intrinsic immunogenicity. Therefore, ApoE may serve as a predictor and a potential therapeutic target for ICB resistance in M2-TAMs-enriched cancer patients. Collectively, our findings signify that the exosome-mediated transfer of functional ApoE from M2 macrophages to the tumor cells confers ICB resistance. Our findings also provide a preclinical rationale for treating M2-enriched tumors with ApoE ligand, EZ-482, to restore sensitivity to ICB immunotherapy.
format Online
Article
Text
id pubmed-10184604
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Taylor & Francis
record_format MEDLINE/PubMed
spelling pubmed-101846042023-05-16 M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance Zheng, Naisheng Wang, Tingting Luo, Qin Liu, Yi Yang, Junyao Zhou, Yunlan Xie, Guohua Ma, Yanhui Yuan, Xiangliang Shen, Lisong Oncoimmunology Research Article T-cell-based immune checkpoint blockade therapy (ICB) can be undermined by local immunosuppressive M2-like tumor-associated macrophages (TAMs). However, modulating macrophages has proved difficult as the molecular and functional features of M2-TAMs on tumor growth are still uncertain. Here we reported that immunosuppressive M2 macrophages render cancer cells resistant to CD8(+) T-cell-dependent tumor-killing refractory ICB efficacy by secreting exosomes. Proteomics and functional studies revealed that M2 macrophage-derived exosome (M2-exo) transmitted apolipoprotein E (ApoE) to cancer cells conferring ICB resistance by downregulated MHC-I expression curbing tumor intrinsic immunogenicity. Mechanistically, M2 exosomal ApoE diminished the tumor-intrinsic ATPase activity of binding immunoglobulin protein (BiP) to decrease tumor MHC-I expression. Sensitizing ICB efficacy can be achieved by the administration of ApoE ligand, EZ-482, enhancing ATPase activity of BiP to boost tumor-intrinsic immunogenicity. Therefore, ApoE may serve as a predictor and a potential therapeutic target for ICB resistance in M2-TAMs-enriched cancer patients. Collectively, our findings signify that the exosome-mediated transfer of functional ApoE from M2 macrophages to the tumor cells confers ICB resistance. Our findings also provide a preclinical rationale for treating M2-enriched tumors with ApoE ligand, EZ-482, to restore sensitivity to ICB immunotherapy. Taylor & Francis 2023-05-13 /pmc/articles/PMC10184604/ /pubmed/37197441 http://dx.doi.org/10.1080/2162402X.2023.2210959 Text en © 2023 The Author(s). Published with license by Taylor & Francis Group, LLC. https://creativecommons.org/licenses/by-nc/4.0/This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) ), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited. The terms on which this article has been published allow the posting of the Accepted Manuscript in a repository by the author(s) or with their consent.
spellingShingle Research Article
Zheng, Naisheng
Wang, Tingting
Luo, Qin
Liu, Yi
Yang, Junyao
Zhou, Yunlan
Xie, Guohua
Ma, Yanhui
Yuan, Xiangliang
Shen, Lisong
M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance
title M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance
title_full M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance
title_fullStr M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance
title_full_unstemmed M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance
title_short M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance
title_sort m2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10184604/
https://www.ncbi.nlm.nih.gov/pubmed/37197441
http://dx.doi.org/10.1080/2162402X.2023.2210959
work_keys_str_mv AT zhengnaisheng m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT wangtingting m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT luoqin m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT liuyi m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT yangjunyao m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT zhouyunlan m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT xieguohua m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT mayanhui m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT yuanxiangliang m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance
AT shenlisong m2macrophagederivedexosomessuppresstumorintrinsicimmunogenicitytoconferimmunotherapyresistance