Cargando…

Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy

Background: Post-surgical recurrence of the metastatic colorectal cancer (mCRC) remains a challenge, even with adjuvant therapy. Moreover, patients show variable outcomes. Here, we set to identify gene models based on the perspectives of intrinsic cell activities and extrinsic immune microenvironmen...

Descripción completa

Detalles Bibliográficos
Autores principales: Luo, Zhiwen, Chen, Xiao, Zhang, Yefan, Huang, Zhen, Zhao, Hong, Zhao, Jianjun, Li, Zhiyu, Zhou, Jianguo, Liu, Jianmei, Cai, Jianqiang, Bi, Xinyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7876250/
https://www.ncbi.nlm.nih.gov/pubmed/33585439
http://dx.doi.org/10.3389/fcell.2020.577125
_version_ 1783649931839406080
author Luo, Zhiwen
Chen, Xiao
Zhang, Yefan
Huang, Zhen
Zhao, Hong
Zhao, Jianjun
Li, Zhiyu
Zhou, Jianguo
Liu, Jianmei
Cai, Jianqiang
Bi, Xinyu
author_facet Luo, Zhiwen
Chen, Xiao
Zhang, Yefan
Huang, Zhen
Zhao, Hong
Zhao, Jianjun
Li, Zhiyu
Zhou, Jianguo
Liu, Jianmei
Cai, Jianqiang
Bi, Xinyu
author_sort Luo, Zhiwen
collection PubMed
description Background: Post-surgical recurrence of the metastatic colorectal cancer (mCRC) remains a challenge, even with adjuvant therapy. Moreover, patients show variable outcomes. Here, we set to identify gene models based on the perspectives of intrinsic cell activities and extrinsic immune microenvironment to predict the recurrence of mCRC and guide the adjuvant therapy. Methods: An RNA-based gene expression analysis of CRC samples (total = 998, including mCRCs = 344, non-mCRCs = 654) was performed. A metastasis-evaluation model (MEM) for mCRCs was developed using the Cox survival model based on the prognostic differentially expressed genes between mCRCs and non-mCRCs. This model separated the mCRC samples into high- and low-recurrence risk clusters that were tested using machine learning to predict recurrence. Further, an immune prognostic model (IPM) was built using the COX survival model with the prognostic differentially expressed immune-related genes between the two MEM risk clusters. The ability of MEM and IPM to predict prognosis was analyzed and validated. Moreover, the IPM was utilized to evaluate its relationship with the immune microenvironment and response to immuno-/chemotherapy. Finally, the dysregulation cause of IPM three genes was analyzed in bioinformatics. Results: A high post-operative recurrence risk was observed owing to the downregulation of the immune response, which was influenced by MEM genes (BAMBI, F13A1, LCN2) and their related IPM genes (SLIT2, CDKN2A, CLU). The MEM and IPM were developed and validated through mCRC samples to differentiate between low- and high-recurrence risk in a real-world cohort. The functional enrichment analysis suggested pathways related to immune response and immune system diseases as the major functional pathways related to the IPM genes. The IPM high-risk group (IPM-high) showed higher fractions of regulatory T cells (Tregs) and smaller fractions of resting memory CD4+ T cells than the IPM-low group. Moreover, the stroma and immune cells in the IPM-high samples were scant. Further, the IPM-high group showed downregulation of MHC class II molecules. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and GDSC analysis suggested the IPM-low as a promising responder to anti-CTLA-4 therapy and the common FDA-targeted drugs, while the IPM-high was non-responsive to these treatments. However, treatment using anti-CDKN2A agents, along with the activation of major histocompatibility complex (MHC) class-II response might sensitize this refractory mCRC subgroup. The dysfunction of MEIS1 might be the reason for the dysregulation of IPM genes. Conclusions: The IPM could identify subgroups of mCRC with a distinct risk of recurrence and stratify the patients sensitive to immuno-/chemotherapy. Further, for the first time, our study highlights the importance of MHC class-II molecules in the treatment of mCRCs using immunotherapy.
format Online
Article
Text
id pubmed-7876250
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-78762502021-02-12 Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy Luo, Zhiwen Chen, Xiao Zhang, Yefan Huang, Zhen Zhao, Hong Zhao, Jianjun Li, Zhiyu Zhou, Jianguo Liu, Jianmei Cai, Jianqiang Bi, Xinyu Front Cell Dev Biol Cell and Developmental Biology Background: Post-surgical recurrence of the metastatic colorectal cancer (mCRC) remains a challenge, even with adjuvant therapy. Moreover, patients show variable outcomes. Here, we set to identify gene models based on the perspectives of intrinsic cell activities and extrinsic immune microenvironment to predict the recurrence of mCRC and guide the adjuvant therapy. Methods: An RNA-based gene expression analysis of CRC samples (total = 998, including mCRCs = 344, non-mCRCs = 654) was performed. A metastasis-evaluation model (MEM) for mCRCs was developed using the Cox survival model based on the prognostic differentially expressed genes between mCRCs and non-mCRCs. This model separated the mCRC samples into high- and low-recurrence risk clusters that were tested using machine learning to predict recurrence. Further, an immune prognostic model (IPM) was built using the COX survival model with the prognostic differentially expressed immune-related genes between the two MEM risk clusters. The ability of MEM and IPM to predict prognosis was analyzed and validated. Moreover, the IPM was utilized to evaluate its relationship with the immune microenvironment and response to immuno-/chemotherapy. Finally, the dysregulation cause of IPM three genes was analyzed in bioinformatics. Results: A high post-operative recurrence risk was observed owing to the downregulation of the immune response, which was influenced by MEM genes (BAMBI, F13A1, LCN2) and their related IPM genes (SLIT2, CDKN2A, CLU). The MEM and IPM were developed and validated through mCRC samples to differentiate between low- and high-recurrence risk in a real-world cohort. The functional enrichment analysis suggested pathways related to immune response and immune system diseases as the major functional pathways related to the IPM genes. The IPM high-risk group (IPM-high) showed higher fractions of regulatory T cells (Tregs) and smaller fractions of resting memory CD4+ T cells than the IPM-low group. Moreover, the stroma and immune cells in the IPM-high samples were scant. Further, the IPM-high group showed downregulation of MHC class II molecules. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and GDSC analysis suggested the IPM-low as a promising responder to anti-CTLA-4 therapy and the common FDA-targeted drugs, while the IPM-high was non-responsive to these treatments. However, treatment using anti-CDKN2A agents, along with the activation of major histocompatibility complex (MHC) class-II response might sensitize this refractory mCRC subgroup. The dysfunction of MEIS1 might be the reason for the dysregulation of IPM genes. Conclusions: The IPM could identify subgroups of mCRC with a distinct risk of recurrence and stratify the patients sensitive to immuno-/chemotherapy. Further, for the first time, our study highlights the importance of MHC class-II molecules in the treatment of mCRCs using immunotherapy. Frontiers Media S.A. 2021-01-28 /pmc/articles/PMC7876250/ /pubmed/33585439 http://dx.doi.org/10.3389/fcell.2020.577125 Text en Copyright © 2021 Luo, Chen, Zhang, Huang, Zhao, Zhao, Li, Zhou, Liu, Cai and Bi. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Cell and Developmental Biology
Luo, Zhiwen
Chen, Xiao
Zhang, Yefan
Huang, Zhen
Zhao, Hong
Zhao, Jianjun
Li, Zhiyu
Zhou, Jianguo
Liu, Jianmei
Cai, Jianqiang
Bi, Xinyu
Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy
title Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy
title_full Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy
title_fullStr Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy
title_full_unstemmed Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy
title_short Development of a Metastasis-Related Immune Prognostic Model of Metastatic Colorectal Cancer and Its Usefulness to Immunotherapy
title_sort development of a metastasis-related immune prognostic model of metastatic colorectal cancer and its usefulness to immunotherapy
topic Cell and Developmental Biology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7876250/
https://www.ncbi.nlm.nih.gov/pubmed/33585439
http://dx.doi.org/10.3389/fcell.2020.577125
work_keys_str_mv AT luozhiwen developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT chenxiao developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT zhangyefan developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT huangzhen developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT zhaohong developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT zhaojianjun developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT lizhiyu developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT zhoujianguo developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT liujianmei developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT caijianqiang developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy
AT bixinyu developmentofametastasisrelatedimmuneprognosticmodelofmetastaticcolorectalcanceranditsusefulnesstoimmunotherapy