Cargando…

Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma

BACKGROUND: Lung squamous cell carcinoma (LUSC) is a highly malignant tumor with an extremely poor prognosis. Immune checkpoint inhibitors (ICIs) improve survival in some patients with LUSC. Tumor mutation burden (TMB) is a useful biomarker to predict the efficacy of ICIs. However, predictive and pr...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Yuting, Xu, Min, Zhao, Kai, Liu, Baodong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10089836/
https://www.ncbi.nlm.nih.gov/pubmed/37065576
http://dx.doi.org/10.21037/jtd-23-103
_version_ 1785022845328293888
author Zhou, Yuting
Xu, Min
Zhao, Kai
Liu, Baodong
author_facet Zhou, Yuting
Xu, Min
Zhao, Kai
Liu, Baodong
author_sort Zhou, Yuting
collection PubMed
description BACKGROUND: Lung squamous cell carcinoma (LUSC) is a highly malignant tumor with an extremely poor prognosis. Immune checkpoint inhibitors (ICIs) improve survival in some patients with LUSC. Tumor mutation burden (TMB) is a useful biomarker to predict the efficacy of ICIs. However, predictive and prognostic factors related to TMB in LUSC remain elusive. This study aimed to find effective biomarkers based on TMB and immune response and establish a prognostic model of LUSC. METHODS: We downloaded Mutation Annotation Format (MAF) files from The Cancer Genome Atlas (TCGA) database and identified immune-related differentially expressed genes (DEGs) between high- and low-TMB groups. The prognostic model was established using cox regression. The primary outcome was overall survival (OS). Receiver operating characteristic (ROC) curves and calibration curves were used to verify the accuracy of the model. GSE37745 acted as external validation set. The expression and prognosis of hub genes as well as their correlation with immune cells and somatic copy number variation (sCNA) were analyzed. RESULTS: The TMB of patients with LUSC was correlated with prognosis and stage. High TMB group had higher survival rate (P<0.001). Five TMB-related hub immune genes (TINAGL1, FGFR2, CTSE, SFTPA1, and IGHV7-81) were identified and the prognostic model was constructed. The survival time of high-risk group was significantly shorter than that of low-risk group (P<0.001). The validation results of the model were quite stable in different data sets, and the area under curve (AUC) of training set and validation set were 0.658 and 0.644, respectively. Calibration chart, risk curve, and nomogram revealed that the prognostic model was reliable in predicting the prognostic risk of LUSC, and the risk score of the model could be used as an independent prognostic factor for LUSC patients (P<0.001). CONCLUSIONS: Our results show that high TMB is associated with poor prognosis in patients with LUSC. The prognostic model related to TMB and immunity can effectively predict the prognosis of LUSC, and risk score is one of the independent prognostic factors of LUSC. However, this study still has some limitations, which need to be further verified in large-scale and prospective studies.
format Online
Article
Text
id pubmed-10089836
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-100898362023-04-13 Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma Zhou, Yuting Xu, Min Zhao, Kai Liu, Baodong J Thorac Dis Original Article BACKGROUND: Lung squamous cell carcinoma (LUSC) is a highly malignant tumor with an extremely poor prognosis. Immune checkpoint inhibitors (ICIs) improve survival in some patients with LUSC. Tumor mutation burden (TMB) is a useful biomarker to predict the efficacy of ICIs. However, predictive and prognostic factors related to TMB in LUSC remain elusive. This study aimed to find effective biomarkers based on TMB and immune response and establish a prognostic model of LUSC. METHODS: We downloaded Mutation Annotation Format (MAF) files from The Cancer Genome Atlas (TCGA) database and identified immune-related differentially expressed genes (DEGs) between high- and low-TMB groups. The prognostic model was established using cox regression. The primary outcome was overall survival (OS). Receiver operating characteristic (ROC) curves and calibration curves were used to verify the accuracy of the model. GSE37745 acted as external validation set. The expression and prognosis of hub genes as well as their correlation with immune cells and somatic copy number variation (sCNA) were analyzed. RESULTS: The TMB of patients with LUSC was correlated with prognosis and stage. High TMB group had higher survival rate (P<0.001). Five TMB-related hub immune genes (TINAGL1, FGFR2, CTSE, SFTPA1, and IGHV7-81) were identified and the prognostic model was constructed. The survival time of high-risk group was significantly shorter than that of low-risk group (P<0.001). The validation results of the model were quite stable in different data sets, and the area under curve (AUC) of training set and validation set were 0.658 and 0.644, respectively. Calibration chart, risk curve, and nomogram revealed that the prognostic model was reliable in predicting the prognostic risk of LUSC, and the risk score of the model could be used as an independent prognostic factor for LUSC patients (P<0.001). CONCLUSIONS: Our results show that high TMB is associated with poor prognosis in patients with LUSC. The prognostic model related to TMB and immunity can effectively predict the prognosis of LUSC, and risk score is one of the independent prognostic factors of LUSC. However, this study still has some limitations, which need to be further verified in large-scale and prospective studies. AME Publishing Company 2023-03-29 2023-03-31 /pmc/articles/PMC10089836/ /pubmed/37065576 http://dx.doi.org/10.21037/jtd-23-103 Text en 2023 Journal of Thoracic Disease. All rights reserved. https://creativecommons.org/licenses/by-nc-nd/4.0/Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0 (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle Original Article
Zhou, Yuting
Xu, Min
Zhao, Kai
Liu, Baodong
Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma
title Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma
title_full Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma
title_fullStr Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma
title_full_unstemmed Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma
title_short Construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma
title_sort construction and validation of a tumor mutational burden and immune-related prognostic model for predicting the prognosis of patients with lung squamous cell carcinoma
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10089836/
https://www.ncbi.nlm.nih.gov/pubmed/37065576
http://dx.doi.org/10.21037/jtd-23-103
work_keys_str_mv AT zhouyuting constructionandvalidationofatumormutationalburdenandimmunerelatedprognosticmodelforpredictingtheprognosisofpatientswithlungsquamouscellcarcinoma
AT xumin constructionandvalidationofatumormutationalburdenandimmunerelatedprognosticmodelforpredictingtheprognosisofpatientswithlungsquamouscellcarcinoma
AT zhaokai constructionandvalidationofatumormutationalburdenandimmunerelatedprognosticmodelforpredictingtheprognosisofpatientswithlungsquamouscellcarcinoma
AT liubaodong constructionandvalidationofatumormutationalburdenandimmunerelatedprognosticmodelforpredictingtheprognosisofpatientswithlungsquamouscellcarcinoma