Cargando…

MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response

BACKGROUND: The efficacy of anti‐programmed cell death protein 1 (PD‐1) immunotherapy in various cancers, including gastric cancer (GC), needs to be potentiated by more effective targeting to enhance therapeutic efficacy or identifying accurate biomarkers to predict clinical responses. Here, we atte...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Bin, Wang, Chun‐Mei, Wu, Hao‐Xiang, Wang, Feng, Chai, Yang‐Yang, Hu, Ye, Wang, Bing‐Jing, Yu, Zhou, Xia, Rong‐Hua, Xu, Rui‐Hua, Cao, Xue‐Tao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10565382/
https://www.ncbi.nlm.nih.gov/pubmed/37539769
http://dx.doi.org/10.1002/cac2.12476
_version_ 1785118683137310720
author Zhang, Bin
Wang, Chun‐Mei
Wu, Hao‐Xiang
Wang, Feng
Chai, Yang‐Yang
Hu, Ye
Wang, Bing‐Jing
Yu, Zhou
Xia, Rong‐Hua
Xu, Rui‐Hua
Cao, Xue‐Tao
author_facet Zhang, Bin
Wang, Chun‐Mei
Wu, Hao‐Xiang
Wang, Feng
Chai, Yang‐Yang
Hu, Ye
Wang, Bing‐Jing
Yu, Zhou
Xia, Rong‐Hua
Xu, Rui‐Hua
Cao, Xue‐Tao
author_sort Zhang, Bin
collection PubMed
description BACKGROUND: The efficacy of anti‐programmed cell death protein 1 (PD‐1) immunotherapy in various cancers, including gastric cancer (GC), needs to be potentiated by more effective targeting to enhance therapeutic efficacy or identifying accurate biomarkers to predict clinical responses. Here, we attempted to identify molecules predicting or/and promoting anti‐PD‐1 therapeutic response in advanced GC (AGC). METHODS: The transcriptome of AGC tissues from patients with different clinical responses to anti‐PD‐1 immunotherapy and GC cells was analyzed by RNA sequencing. The protein and mRNA levels of the major facilitator superfamily domain containing 2A (MFSD2A) in GC cells were assessed via quantitative real‐time polymerase chain reaction, Western blotting, and immunohistochemistry. Additionally, the regulation of anti‐PD‐1 response by MFSD2A was studied in tumor‐bearing mice. Cytometry by Time‐of‐Flight, multiple immunohistochemistry, and flow cytometry assays were used to explore immunological responses. The effects of MFSD2A on lipid metabolism in mice cancer tissue and GC cells was detected by metabolomics. RESULTS: Higher expression of MFSD2A in tumor tissues of AGC patients was associated with better response to anti‐PD‐1 immunotherapy. Moreover, MFSD2A expression was lower in GC tissues compared to adjacent normal tissues, and its expression was inversely correlated with GC stage. The overexpression of MFSD2A in GC cells enhanced the efficacy of anti‐PD‐1 immunotherapy in vivo by reprogramming the tumor microenvironment (TME), characterized by increased CD8(+) T cell activation and reduced its exhaustion. MFSD2A inhibited transforming growth factor β1 (TGFβ1) release from GC cells by suppressing cyclooxygenase 2 (COX2)‐prostaglandin synthesis, which consequently reprogrammed TME to promote anti‐tumor T cell activation. CONCLUSIONS: MFSD2A potentially serves as a predictive biomarker for anti‐PD‐1 immunotherapy response in AGC patients. MFSD2A may be a promising therapeutic target to potentiate the efficacy of anti‐PD‐1 immunotherapy by reprogramming the TME to promote T cells activation.
format Online
Article
Text
id pubmed-10565382
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-105653822023-10-12 MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response Zhang, Bin Wang, Chun‐Mei Wu, Hao‐Xiang Wang, Feng Chai, Yang‐Yang Hu, Ye Wang, Bing‐Jing Yu, Zhou Xia, Rong‐Hua Xu, Rui‐Hua Cao, Xue‐Tao Cancer Commun (Lond) Original Articles BACKGROUND: The efficacy of anti‐programmed cell death protein 1 (PD‐1) immunotherapy in various cancers, including gastric cancer (GC), needs to be potentiated by more effective targeting to enhance therapeutic efficacy or identifying accurate biomarkers to predict clinical responses. Here, we attempted to identify molecules predicting or/and promoting anti‐PD‐1 therapeutic response in advanced GC (AGC). METHODS: The transcriptome of AGC tissues from patients with different clinical responses to anti‐PD‐1 immunotherapy and GC cells was analyzed by RNA sequencing. The protein and mRNA levels of the major facilitator superfamily domain containing 2A (MFSD2A) in GC cells were assessed via quantitative real‐time polymerase chain reaction, Western blotting, and immunohistochemistry. Additionally, the regulation of anti‐PD‐1 response by MFSD2A was studied in tumor‐bearing mice. Cytometry by Time‐of‐Flight, multiple immunohistochemistry, and flow cytometry assays were used to explore immunological responses. The effects of MFSD2A on lipid metabolism in mice cancer tissue and GC cells was detected by metabolomics. RESULTS: Higher expression of MFSD2A in tumor tissues of AGC patients was associated with better response to anti‐PD‐1 immunotherapy. Moreover, MFSD2A expression was lower in GC tissues compared to adjacent normal tissues, and its expression was inversely correlated with GC stage. The overexpression of MFSD2A in GC cells enhanced the efficacy of anti‐PD‐1 immunotherapy in vivo by reprogramming the tumor microenvironment (TME), characterized by increased CD8(+) T cell activation and reduced its exhaustion. MFSD2A inhibited transforming growth factor β1 (TGFβ1) release from GC cells by suppressing cyclooxygenase 2 (COX2)‐prostaglandin synthesis, which consequently reprogrammed TME to promote anti‐tumor T cell activation. CONCLUSIONS: MFSD2A potentially serves as a predictive biomarker for anti‐PD‐1 immunotherapy response in AGC patients. MFSD2A may be a promising therapeutic target to potentiate the efficacy of anti‐PD‐1 immunotherapy by reprogramming the TME to promote T cells activation. John Wiley and Sons Inc. 2023-08-04 /pmc/articles/PMC10565382/ /pubmed/37539769 http://dx.doi.org/10.1002/cac2.12476 Text en © 2023 The Authors. Cancer Communications published by John Wiley & Sons Australia, Ltd. on behalf of Sun Yat‐sen University Cancer Center. 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
Zhang, Bin
Wang, Chun‐Mei
Wu, Hao‐Xiang
Wang, Feng
Chai, Yang‐Yang
Hu, Ye
Wang, Bing‐Jing
Yu, Zhou
Xia, Rong‐Hua
Xu, Rui‐Hua
Cao, Xue‐Tao
MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response
title MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response
title_full MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response
title_fullStr MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response
title_full_unstemmed MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response
title_short MFSD2A potentiates gastric cancer response to anti‐PD‐1 immunotherapy by reprogramming the tumor microenvironment to activate T cell response
title_sort mfsd2a potentiates gastric cancer response to anti‐pd‐1 immunotherapy by reprogramming the tumor microenvironment to activate t cell response
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10565382/
https://www.ncbi.nlm.nih.gov/pubmed/37539769
http://dx.doi.org/10.1002/cac2.12476
work_keys_str_mv AT zhangbin mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT wangchunmei mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT wuhaoxiang mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT wangfeng mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT chaiyangyang mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT huye mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT wangbingjing mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT yuzhou mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT xiaronghua mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT xuruihua mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse
AT caoxuetao mfsd2apotentiatesgastriccancerresponsetoantipd1immunotherapybyreprogrammingthetumormicroenvironmenttoactivatetcellresponse