Cargando…

FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts

Background: Since T cell exclusion contributes to tumor immune evasion and immunotherapy resistance, how to improve T cell infiltration into solid tumors becomes an urgent challenge. Methods: We employed deep learning to profile the tumor immune microenvironment (TIME) in triple negative breast canc...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Yushen, Yi, Ziying, Li, Jie, Wei, Yuxian, Feng, Rui, Liu, Jiazhou, Huang, Jiefeng, Chen, Yuru, Wang, Xiaoyu, Sun, Jiazheng, Yin, Xuedong, Li, Yunhai, Wan, Jingyuan, Zhang, Li, Huang, Jing, Du, Huimin, Wang, Xiaoyi, Li, Qin, Ren, Guosheng, Li, Hongzhong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9254240/
https://www.ncbi.nlm.nih.gov/pubmed/35832090
http://dx.doi.org/10.7150/thno.68972
_version_ 1784740652924272640
author Wu, Yushen
Yi, Ziying
Li, Jie
Wei, Yuxian
Feng, Rui
Liu, Jiazhou
Huang, Jiefeng
Chen, Yuru
Wang, Xiaoyu
Sun, Jiazheng
Yin, Xuedong
Li, Yunhai
Wan, Jingyuan
Zhang, Li
Huang, Jing
Du, Huimin
Wang, Xiaoyi
Li, Qin
Ren, Guosheng
Li, Hongzhong
author_facet Wu, Yushen
Yi, Ziying
Li, Jie
Wei, Yuxian
Feng, Rui
Liu, Jiazhou
Huang, Jiefeng
Chen, Yuru
Wang, Xiaoyu
Sun, Jiazheng
Yin, Xuedong
Li, Yunhai
Wan, Jingyuan
Zhang, Li
Huang, Jing
Du, Huimin
Wang, Xiaoyi
Li, Qin
Ren, Guosheng
Li, Hongzhong
author_sort Wu, Yushen
collection PubMed
description Background: Since T cell exclusion contributes to tumor immune evasion and immunotherapy resistance, how to improve T cell infiltration into solid tumors becomes an urgent challenge. Methods: We employed deep learning to profile the tumor immune microenvironment (TIME) in triple negative breast cancer (TNBC) samples from TCGA datasets and noticed that fibroblast growth factor receptor (FGFR) signaling pathways were enriched in the immune-excluded phenotype of TNBC. Erdafitinib, a selective FGFR inhibitor, was then used to investigate the effect of FGFR blockade on TIME landscape of TNBC syngeneic mouse models by flow cytometry, mass cytometry (CyTOF) and RNA sequencing. Cell Counting Kit-8 (CCK-8) assay and transwell migration assay were carried out to detect the effect of FGFR blockade on cell proliferation and migration, respectively. Cytokine array, western blot, enzyme-linked immunosorbent assay (ELISA) and immunofluorescence (IF) were employed to investigate the potential mechanism by which FGFR inhibition enhanced T cell infiltration. Results: Blocking FGFR pathway by Erdafitinib markedly suppressed tumor growth with increased T cell infiltration in immunocompetent mouse models of TNBC. Mechanistically, FGFR blockade inhibited cancer-associated fibroblasts (CAFs) proliferation, migration and secretion of vascular cell adhesion molecule 1 (VCAM-1) by down-regulating MAPK/ERK pathway in CAFs, thus promoting T cell infiltration by breaking physical and chemical barriers built by CAFs in TIME. Furthermore, we observed that FGFR inhibition combined with immune checkpoint blockade therapy (ICT) greatly improved the therapeutic response of TNBC tumor models. Conclusions: FGFR blockade enhanced ICT response by turning immune “cold” tumor into “hot” tumor, providing remarkable implications of FGFR inhibitors as adjuvant agents for combinatorial immunotherapy.
format Online
Article
Text
id pubmed-9254240
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-92542402022-07-12 FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts Wu, Yushen Yi, Ziying Li, Jie Wei, Yuxian Feng, Rui Liu, Jiazhou Huang, Jiefeng Chen, Yuru Wang, Xiaoyu Sun, Jiazheng Yin, Xuedong Li, Yunhai Wan, Jingyuan Zhang, Li Huang, Jing Du, Huimin Wang, Xiaoyi Li, Qin Ren, Guosheng Li, Hongzhong Theranostics Research Paper Background: Since T cell exclusion contributes to tumor immune evasion and immunotherapy resistance, how to improve T cell infiltration into solid tumors becomes an urgent challenge. Methods: We employed deep learning to profile the tumor immune microenvironment (TIME) in triple negative breast cancer (TNBC) samples from TCGA datasets and noticed that fibroblast growth factor receptor (FGFR) signaling pathways were enriched in the immune-excluded phenotype of TNBC. Erdafitinib, a selective FGFR inhibitor, was then used to investigate the effect of FGFR blockade on TIME landscape of TNBC syngeneic mouse models by flow cytometry, mass cytometry (CyTOF) and RNA sequencing. Cell Counting Kit-8 (CCK-8) assay and transwell migration assay were carried out to detect the effect of FGFR blockade on cell proliferation and migration, respectively. Cytokine array, western blot, enzyme-linked immunosorbent assay (ELISA) and immunofluorescence (IF) were employed to investigate the potential mechanism by which FGFR inhibition enhanced T cell infiltration. Results: Blocking FGFR pathway by Erdafitinib markedly suppressed tumor growth with increased T cell infiltration in immunocompetent mouse models of TNBC. Mechanistically, FGFR blockade inhibited cancer-associated fibroblasts (CAFs) proliferation, migration and secretion of vascular cell adhesion molecule 1 (VCAM-1) by down-regulating MAPK/ERK pathway in CAFs, thus promoting T cell infiltration by breaking physical and chemical barriers built by CAFs in TIME. Furthermore, we observed that FGFR inhibition combined with immune checkpoint blockade therapy (ICT) greatly improved the therapeutic response of TNBC tumor models. Conclusions: FGFR blockade enhanced ICT response by turning immune “cold” tumor into “hot” tumor, providing remarkable implications of FGFR inhibitors as adjuvant agents for combinatorial immunotherapy. Ivyspring International Publisher 2022-05-27 /pmc/articles/PMC9254240/ /pubmed/35832090 http://dx.doi.org/10.7150/thno.68972 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Wu, Yushen
Yi, Ziying
Li, Jie
Wei, Yuxian
Feng, Rui
Liu, Jiazhou
Huang, Jiefeng
Chen, Yuru
Wang, Xiaoyu
Sun, Jiazheng
Yin, Xuedong
Li, Yunhai
Wan, Jingyuan
Zhang, Li
Huang, Jing
Du, Huimin
Wang, Xiaoyi
Li, Qin
Ren, Guosheng
Li, Hongzhong
FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
title FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
title_full FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
title_fullStr FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
title_full_unstemmed FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
title_short FGFR blockade boosts T cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
title_sort fgfr blockade boosts t cell infiltration into triple-negative breast cancer by regulating cancer-associated fibroblasts
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9254240/
https://www.ncbi.nlm.nih.gov/pubmed/35832090
http://dx.doi.org/10.7150/thno.68972
work_keys_str_mv AT wuyushen fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT yiziying fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT lijie fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT weiyuxian fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT fengrui fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT liujiazhou fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT huangjiefeng fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT chenyuru fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT wangxiaoyu fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT sunjiazheng fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT yinxuedong fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT liyunhai fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT wanjingyuan fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT zhangli fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT huangjing fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT duhuimin fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT wangxiaoyi fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT liqin fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT renguosheng fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts
AT lihongzhong fgfrblockadebooststcellinfiltrationintotriplenegativebreastcancerbyregulatingcancerassociatedfibroblasts