File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Exosomes derived from 3δ -T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment

TitleExosomes derived from 3δ -T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment
Authors
Keywordshead and neck neoplasms
immunotherapy
radiotherapy
t-lymphocytes
tumor microenvironment
Issue Date2022
Citation
Journal for ImmunoTherapy of Cancer, 2022, v. 10, n. 2, article no. e003832 How to Cite?
AbstractBackground Radiotherapy is the first-line treatment for patients nasopharyngeal carcinoma (NPC), but its therapeutic efficacy is poor in some patients due to radioresistance. Adoptive T cell-based immunotherapy has also shown promise to control NPC; however, its antitumor efficacy may be attenuated by an immunosuppressive tumor microenvironment. Exosomes derived from 3δ -T cells ( 3δ -T-Exos) have potent antitumor potentials. However, it remains unknown whether 3δ -T-Exos have synergistic effect with radiotherapy and preserve their antitumor activities against NPC in an immunosuppressive tumor microenvironment. Methods 3δ -T-Exos were stained with fluorescent membrane dye, and their interactions with NPC were determined both in vitro and in vivo. NPC cell deaths were detected after treatment with 3δ -T-Exos and/or irradiation. Moreover, effects of 3δ -T-Exos on radioresistant cancer stem-like cells (CSCs) were determined. The therapeutic efficacy of combination therapy using 3δ -T-Exos and irradiation on NPC tumor progression was also monitored in vivo. Finally, the tumor-killing and T cell-promoting activities of 3δ -T-Exos were determined under the culture in immunosuppressive NPC supernatant. Results 3δ -T-Exos effectively interacted with NPC tumor cells in vitro and in vivo. 3δ -T-Exos not only killed NPC cells in vitro, which was mainly mediated by Fas/Fas ligand (FasL) and death receptor 5 (DR5)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathways, but also controlled NPC tumor growth and prolonged tumor-bearing mice survival in vivo. Furthermore, 3δ -T-Exos selectively targeted the radioresistant CD44 +/high CSCs and induced profound cell apoptosis. The combination of 3δ -T-Exos with radiotherapy overcame the radioresistance of CD44 +/high NPC cells and significantly improved its therapeutic efficacy against NPC in vitro and in vivo. In addition, 3δ -T-Exos promoted T-cell migration into NPC tumors by upregulating CCR5 on T cells that were chemoattracted by CCR5 ligands in the NPC tumor microenvironment. Although NPC tumor cells secreted abundant tumor growth factor beta to suppress T-cell responses, 3δ -T-Exos preserved their direct antitumor activities and overcame the immunosuppressive NPC microenvironment to amplify T-cell antitumor immunity. Conclusions 3δ -T-Exos synergized with radiotherapy to control NPC by overcoming the radioresistance of NPC CSCs. Moreover, 3δ -T-Exos preserved their tumor-killing and T cell-promoting activities in the immunosuppressive NPC microenvironment. This study provides a proof of concept for a novel and potent strategy by combining 3δ -T-Exos with radiotherapy in the control of NPC.
Persistent Identifierhttp://hdl.handle.net/10722/343360
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorWang, Xiwei-
dc.contributor.authorZhang, Yanmei-
dc.contributor.authorMu, Xiaofeng-
dc.contributor.authorTu, Chloe Ran-
dc.contributor.authorChung, Yuet-
dc.contributor.authorTsao, Sai Wah-
dc.contributor.authorChan, Godfrey Chi Fung-
dc.contributor.authorLeung, Wing Hang-
dc.contributor.authorLau, Yu Lung-
dc.contributor.authorLiu, Yinping-
dc.contributor.authorTu, Wenwei-
dc.date.accessioned2024-05-10T09:07:28Z-
dc.date.available2024-05-10T09:07:28Z-
dc.date.issued2022-
dc.identifier.citationJournal for ImmunoTherapy of Cancer, 2022, v. 10, n. 2, article no. e003832-
dc.identifier.urihttp://hdl.handle.net/10722/343360-
dc.description.abstractBackground Radiotherapy is the first-line treatment for patients nasopharyngeal carcinoma (NPC), but its therapeutic efficacy is poor in some patients due to radioresistance. Adoptive T cell-based immunotherapy has also shown promise to control NPC; however, its antitumor efficacy may be attenuated by an immunosuppressive tumor microenvironment. Exosomes derived from 3δ -T cells ( 3δ -T-Exos) have potent antitumor potentials. However, it remains unknown whether 3δ -T-Exos have synergistic effect with radiotherapy and preserve their antitumor activities against NPC in an immunosuppressive tumor microenvironment. Methods 3δ -T-Exos were stained with fluorescent membrane dye, and their interactions with NPC were determined both in vitro and in vivo. NPC cell deaths were detected after treatment with 3δ -T-Exos and/or irradiation. Moreover, effects of 3δ -T-Exos on radioresistant cancer stem-like cells (CSCs) were determined. The therapeutic efficacy of combination therapy using 3δ -T-Exos and irradiation on NPC tumor progression was also monitored in vivo. Finally, the tumor-killing and T cell-promoting activities of 3δ -T-Exos were determined under the culture in immunosuppressive NPC supernatant. Results 3δ -T-Exos effectively interacted with NPC tumor cells in vitro and in vivo. 3δ -T-Exos not only killed NPC cells in vitro, which was mainly mediated by Fas/Fas ligand (FasL) and death receptor 5 (DR5)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathways, but also controlled NPC tumor growth and prolonged tumor-bearing mice survival in vivo. Furthermore, 3δ -T-Exos selectively targeted the radioresistant CD44 +/high CSCs and induced profound cell apoptosis. The combination of 3δ -T-Exos with radiotherapy overcame the radioresistance of CD44 +/high NPC cells and significantly improved its therapeutic efficacy against NPC in vitro and in vivo. In addition, 3δ -T-Exos promoted T-cell migration into NPC tumors by upregulating CCR5 on T cells that were chemoattracted by CCR5 ligands in the NPC tumor microenvironment. Although NPC tumor cells secreted abundant tumor growth factor beta to suppress T-cell responses, 3δ -T-Exos preserved their direct antitumor activities and overcame the immunosuppressive NPC microenvironment to amplify T-cell antitumor immunity. Conclusions 3δ -T-Exos synergized with radiotherapy to control NPC by overcoming the radioresistance of NPC CSCs. Moreover, 3δ -T-Exos preserved their tumor-killing and T cell-promoting activities in the immunosuppressive NPC microenvironment. This study provides a proof of concept for a novel and potent strategy by combining 3δ -T-Exos with radiotherapy in the control of NPC.-
dc.languageeng-
dc.relation.ispartofJournal for ImmunoTherapy of Cancer-
dc.subjecthead and neck neoplasms-
dc.subjectimmunotherapy-
dc.subjectradiotherapy-
dc.subjectt-lymphocytes-
dc.subjecttumor microenvironment-
dc.titleExosomes derived from 3δ -T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment-
dc.typeArticle-
dc.description.naturelink_to_subscribed_fulltext-
dc.identifier.doi10.1136/jitc-2021-003832-
dc.identifier.pmid35105688-
dc.identifier.scopuseid_2-s2.0-85123973901-
dc.identifier.volume10-
dc.identifier.issue2-
dc.identifier.spagearticle no. e003832-
dc.identifier.epagearticle no. e003832-
dc.identifier.eissn2051-1426-
dc.identifier.isiWOS:000750204000002-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats