Cargando…

A prognostic 11-DNA methylation signature for lung squamous cell carcinoma

BACKGROUND: Lung squamous cell carcinoma (LUSC), as the second frequent subtype of lung cancer, causes lots of mortalities primarily due to a lack of precise prognostic markers and timely treatment intervention. Previous studies have constructed several risk prognostic models based on DNA methylatio...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Jianlei, Luo, Liyun, Dong, Jing, Liu, Meijun, Zhai, Dongfeng, Huang, Danqing, Ling, Li, Jia, Xiaoting, Luo, Kai, Zheng, Guopei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7330303/
https://www.ncbi.nlm.nih.gov/pubmed/32642165
http://dx.doi.org/10.21037/jtd.2020.03.31
_version_ 1783553089084588032
author Zhang, Jianlei
Luo, Liyun
Dong, Jing
Liu, Meijun
Zhai, Dongfeng
Huang, Danqing
Ling, Li
Jia, Xiaoting
Luo, Kai
Zheng, Guopei
author_facet Zhang, Jianlei
Luo, Liyun
Dong, Jing
Liu, Meijun
Zhai, Dongfeng
Huang, Danqing
Ling, Li
Jia, Xiaoting
Luo, Kai
Zheng, Guopei
author_sort Zhang, Jianlei
collection PubMed
description BACKGROUND: Lung squamous cell carcinoma (LUSC), as the second frequent subtype of lung cancer, causes lots of mortalities primarily due to a lack of precise prognostic markers and timely treatment intervention. Previous studies have constructed several risk prognostic models based on DNA methylation sites in multiple tumors, whereas, DNA methylation signature of LUSC remains to be built, and its predictive value need to be evaluated. METHODS: The genome-wide DNA methylation data of LUSC samples was obtained from The Cancer Genome Atlas dataset. Univariate Cox analysis and the least absolute shrinkage and selection operator (LASSO) were implemented to identify DNA methylation sites related to overall survival of LUSC patients. Thus, we performed multivariate Cox regression to establish a DNA methylation signature. The Kaplan-Meier (K-M) survival curves and time-dependent receiver operating characteristic (ROC) curves were plotted to estimate the prognostic power of the signature. Comparison with other known prognostic biomarkers, our DNA methylation signature showed higher predictive specificity and sensitivity. In addition, multivariate Cox regression screened out independent prognostic factors and constructed a nomogram. RESULTS: Several statistical methods were performed to construct an 11-DNA methylation signature. LUSC patients were divided into low- and high-risk group based on risk score, and high-risk group had a shorter survival time. According to the results of K-M and ROC analyses, the 11-DNA methylation signature showed significant sensitivity and specificity in predicting the LUSC patients’ overall survival. Finally, we integrated some independent prognostic factors (risk score, metastasis stage, and tobacco smoking history) to construct a nomogram, which has excellent prognostic power and may provide guidance for the therapeutic strategies. CONCLUSIONS: We constructed the first risk prognosis model based on DNA methylation site in LUSC, which showed better predictive ability. In addition, a nomogram integrating the DNA methylation signature, metastasis stage, and tobacco smoking history was developed.
format Online
Article
Text
id pubmed-7330303
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-73303032020-07-07 A prognostic 11-DNA methylation signature for lung squamous cell carcinoma Zhang, Jianlei Luo, Liyun Dong, Jing Liu, Meijun Zhai, Dongfeng Huang, Danqing Ling, Li Jia, Xiaoting Luo, Kai Zheng, Guopei J Thorac Dis Original Article BACKGROUND: Lung squamous cell carcinoma (LUSC), as the second frequent subtype of lung cancer, causes lots of mortalities primarily due to a lack of precise prognostic markers and timely treatment intervention. Previous studies have constructed several risk prognostic models based on DNA methylation sites in multiple tumors, whereas, DNA methylation signature of LUSC remains to be built, and its predictive value need to be evaluated. METHODS: The genome-wide DNA methylation data of LUSC samples was obtained from The Cancer Genome Atlas dataset. Univariate Cox analysis and the least absolute shrinkage and selection operator (LASSO) were implemented to identify DNA methylation sites related to overall survival of LUSC patients. Thus, we performed multivariate Cox regression to establish a DNA methylation signature. The Kaplan-Meier (K-M) survival curves and time-dependent receiver operating characteristic (ROC) curves were plotted to estimate the prognostic power of the signature. Comparison with other known prognostic biomarkers, our DNA methylation signature showed higher predictive specificity and sensitivity. In addition, multivariate Cox regression screened out independent prognostic factors and constructed a nomogram. RESULTS: Several statistical methods were performed to construct an 11-DNA methylation signature. LUSC patients were divided into low- and high-risk group based on risk score, and high-risk group had a shorter survival time. According to the results of K-M and ROC analyses, the 11-DNA methylation signature showed significant sensitivity and specificity in predicting the LUSC patients’ overall survival. Finally, we integrated some independent prognostic factors (risk score, metastasis stage, and tobacco smoking history) to construct a nomogram, which has excellent prognostic power and may provide guidance for the therapeutic strategies. CONCLUSIONS: We constructed the first risk prognosis model based on DNA methylation site in LUSC, which showed better predictive ability. In addition, a nomogram integrating the DNA methylation signature, metastasis stage, and tobacco smoking history was developed. AME Publishing Company 2020-05 /pmc/articles/PMC7330303/ /pubmed/32642165 http://dx.doi.org/10.21037/jtd.2020.03.31 Text en 2020 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
Zhang, Jianlei
Luo, Liyun
Dong, Jing
Liu, Meijun
Zhai, Dongfeng
Huang, Danqing
Ling, Li
Jia, Xiaoting
Luo, Kai
Zheng, Guopei
A prognostic 11-DNA methylation signature for lung squamous cell carcinoma
title A prognostic 11-DNA methylation signature for lung squamous cell carcinoma
title_full A prognostic 11-DNA methylation signature for lung squamous cell carcinoma
title_fullStr A prognostic 11-DNA methylation signature for lung squamous cell carcinoma
title_full_unstemmed A prognostic 11-DNA methylation signature for lung squamous cell carcinoma
title_short A prognostic 11-DNA methylation signature for lung squamous cell carcinoma
title_sort prognostic 11-dna methylation signature for lung squamous cell carcinoma
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7330303/
https://www.ncbi.nlm.nih.gov/pubmed/32642165
http://dx.doi.org/10.21037/jtd.2020.03.31
work_keys_str_mv AT zhangjianlei aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT luoliyun aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT dongjing aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT liumeijun aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT zhaidongfeng aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT huangdanqing aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT lingli aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT jiaxiaoting aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT luokai aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT zhengguopei aprognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT zhangjianlei prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT luoliyun prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT dongjing prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT liumeijun prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT zhaidongfeng prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT huangdanqing prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT lingli prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT jiaxiaoting prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT luokai prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma
AT zhengguopei prognostic11dnamethylationsignatureforlungsquamouscellcarcinoma