Skip to main content
Top

01-12-2021 | Esophageal Cancer | Research article

Discovery and validation of methylation signatures in circulating cell-free DNA for early detection of esophageal cancer: a case-control study

Authors: Guibin Qiao, Weitao Zhuang, Bo Dong, Chengcheng Li, Jiayue Xu, Guoqiang Wang, Liang Xie, Zihao Zhou, Dan Tian, Gang Chen, Jiming Tang, Haiyu Zhou, Dongkun Zhang, Ruiqing Shi, Rixin Chen, Weiqi Nian, Yuzi Zhang, Jing Zhao, Xiaofang Wen, Yu Xu, Bingsi Li, Zhihong Zhang, Shangli Cai, Xiaosong Ben, Yu Qi

Published in: BMC Medicine | Issue 1/2021

Login to get access

Abstract

Background

Plasma cell-free DNA (cfDNA) methylation has shown promising results in the early detection of multiple cancers recently. Here, we conducted a study to investigate the performance of cfDNA methylation in the early detection of esophageal cancer (ESCA).

Methods

Specific methylation markers for ESCA were identified and optimized based on esophageal tumor and paired adjacent tissues (n = 24). Age-matched participants with ESCA (n = 85), benign esophageal diseases (n = 10), and healthy controls (n = 125) were randomized into the training and test sets to develop a classifier to differentiate ESCA from healthy controls and benign esophageal disease. The classifier was further validated in an independent plasma cohort of ESCA patients (n = 83) and healthy controls (n = 98).

Results

In total, 921 differentially methylated regions (DMRs) between tumor and adjacent tissues were identified. The early detection classifier based on those DMRs was first developed and tested in plasma samples, discriminating ESCA patients from benign and healthy controls with a sensitivity of 76.2% (60.5–87.9%) and a specificity of 94.1% (85.7–98.4%) in the test set. The performance of the classifier was consistent irrespective of sex, age, and pathological diagnosis (P > 0.05). In the independent plasma validation cohort, similar performance was observed with a sensitivity of 74.7% (64.0–83.6%) and a specificity of 95.9% (89.9–98.9%). Sensitivity for stage 0–II was 58.8% (44.2–72.4%).

Conclusion

We demonstrated that the cfDNA methylation patterns could distinguish ESCAs from healthy individuals and benign esophageal diseases with promising sensitivity and specificity. Further prospective evaluation of the classifier in the early detection of ESCAs in high-risk individuals is warranted.
Appendix
Available only for authorised users
Literature
1.
go back to reference Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a cancer journal for clinicians. 2018;68(6):394–424. PubMed PMID: 30207593. https://doi.org/10.3322/caac.21492.CrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a cancer journal for clinicians. 2018;68(6):394–424. PubMed PMID: 30207593. https://​doi.​org/​10.​3322/​caac.​21492.CrossRef
4.
go back to reference Wei WQ, Chen ZF, He YT, Feng H, Hou J, Lin DM, et al. Long-term follow-up of a community assignment, one-time endoscopic screening study of esophageal cancer in China. J Clin Oncol. 2015;33(17):1951-7. PubMed PMID: 25940715. Pubmed Central PMCID: PMC4881309 online at www.jco.org. Author contributions are found at the end of this article. Epub 2015/05/06. eng. Wei WQ, Chen ZF, He YT, Feng H, Hou J, Lin DM, et al. Long-term follow-up of a community assignment, one-time endoscopic screening study of esophageal cancer in China. J Clin Oncol. 2015;33(17):1951-7. PubMed PMID: 25940715. Pubmed Central PMCID: PMC4881309 online at www.​jco.​org. Author contributions are found at the end of this article. Epub 2015/05/06. eng.
5.
go back to reference Gupta N, Bansal A, Wani SB, Gaddam S, Rastogi A, Sharma P. Endoscopy for upper GI cancer screening in the general population: a cost-utility analysis. Gastrointestinal Endoscopy. 2011;74(3):610–24 e2.CrossRef Gupta N, Bansal A, Wani SB, Gaddam S, Rastogi A, Sharma P. Endoscopy for upper GI cancer screening in the general population: a cost-utility analysis. Gastrointestinal Endoscopy. 2011;74(3):610–24 e2.CrossRef
8.
go back to reference Soto J, Rodriguez-Antolin C, Vallespín E, de Castro CJ, Ibanez de Caceres I. The impact of next-generation sequencing on the DNA methylation-based translational cancer research. Transl Res. 2016;169:1–18 e1. PubMed PMID: 26687736. Epub 2015/12/22. eng.CrossRef Soto J, Rodriguez-Antolin C, Vallespín E, de Castro CJ, Ibanez de Caceres I. The impact of next-generation sequencing on the DNA methylation-based translational cancer research. Transl Res. 2016;169:1–18 e1. PubMed PMID: 26687736. Epub 2015/12/22. eng.CrossRef
10.
go back to reference Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, et al. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Science Transl Med. 2020;12(524):eaax7533. Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, et al. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Science Transl Med. 2020;12(524):eaax7533.
14.
go back to reference Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan JB, Gao Y, Deconde R, Chen M, Rajapakse I, Friend S, Ideker T, Zhang K Genome-wide methylation profiles reveal quantitative views of human aging rates. Molecular Cell. 2013;49(2):359-367. PubMed PMID: 23177740. Pubmed Central PMCID: 3780611, DOI: https://doi.org/10.1016/j.molcel.2012.10.016. Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan JB, Gao Y, Deconde R, Chen M, Rajapakse I, Friend S, Ideker T, Zhang K Genome-wide methylation profiles reveal quantitative views of human aging rates. Molecular Cell. 2013;49(2):359-367. PubMed PMID: 23177740. Pubmed Central PMCID: 3780611, DOI: https://​doi.​org/​10.​1016/​j.​molcel.​2012.​10.​016.
16.
go back to reference Qiang Gao BL, Shangli Cai, Heng Zhao, Wei Zhang, Ning Li, Weilin Wang, Guibin Qiao, Hao Liu, Zhihong Zhang, Jia Fan. Early detection and localization of multiple cancers using a blood-based methylation assay (ELSA-seq). ESMO Asia Virtual Congress 2020 2020. 20-22 November. Qiang Gao BL, Shangli Cai, Heng Zhao, Wei Zhang, Ning Li, Weilin Wang, Guibin Qiao, Hao Liu, Zhihong Zhang, Jia Fan. Early detection and localization of multiple cancers using a blood-based methylation assay (ELSA-seq). ESMO Asia Virtual Congress 2020 2020. 20-22 November.
17.
go back to reference Mao X, Zhang Z, Zheng X, Xie F, Duan F, Jiang L, et al. Capture-based targeted ultradeep sequencing in paired tissue and plasma samples demonstrates differential subclonal ctDNA-releasing capability in advanced lung cancer. J Thoracic Oncol. 2017;12(4):663–72.CrossRef Mao X, Zhang Z, Zheng X, Xie F, Duan F, Jiang L, et al. Capture-based targeted ultradeep sequencing in paired tissue and plasma samples demonstrates differential subclonal ctDNA-releasing capability in advanced lung cancer. J Thoracic Oncol. 2017;12(4):663–72.CrossRef
18.
go back to reference Yang L, Zhang J, Yang G, Xu H, Lin J, Shao L, et al. The prognostic value of a Methylome-based Malignancy Density Scoring System to predict recurrence risk in early-stage lung adenocarcinoma. Theranostics. 2020;10:7635–44.CrossRef Yang L, Zhang J, Yang G, Xu H, Lin J, Shao L, et al. The prognostic value of a Methylome-based Malignancy Density Scoring System to predict recurrence risk in early-stage lung adenocarcinoma. Theranostics. 2020;10:7635–44.CrossRef
19.
go back to reference Guo S, Diep D, Plongthongkum N, Fung HL, Zhang K, Zhang K. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet. 2017;49(4):635–42. PubMed PMID: 28263317. Pubmed Central PMCID: PMC5374016. Epub 2017/03/07. eng. https://doi.org/10.1038/ng.3805.CrossRefPubMedPubMedCentral Guo S, Diep D, Plongthongkum N, Fung HL, Zhang K, Zhang K. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet. 2017;49(4):635–42. PubMed PMID: 28263317. Pubmed Central PMCID: PMC5374016. Epub 2017/03/07. eng. https://​doi.​org/​10.​1038/​ng.​3805.CrossRefPubMedPubMedCentral
21.
go back to reference Pedregosa F, Varoquaux G, Gramfort A, Michel V, Thirion B, Grisel O, et al. Scikit-learn: machine learning in Python. J Mach Learn Res. 2011;12(null):2825–30. Pedregosa F, Varoquaux G, Gramfort A, Michel V, Thirion B, Grisel O, et al. Scikit-learn: machine learning in Python. J Mach Learn Res. 2011;12(null):2825–30.
22.
go back to reference Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature Protocols. 2009;4(1):44.CrossRef Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature Protocols. 2009;4(1):44.CrossRef
27.
go back to reference Gao Q, Li B, Cai S, Xu J, Wang C, Su J, et al. Early detection and localization of multiple cancers using a blood-based methylation assay (ELSA-seq). J Clin Oncol. 2021;39(3_suppl):459.CrossRef Gao Q, Li B, Cai S, Xu J, Wang C, Su J, et al. Early detection and localization of multiple cancers using a blood-based methylation assay (ELSA-seq). J Clin Oncol. 2021;39(3_suppl):459.CrossRef
Metadata
Title
Discovery and validation of methylation signatures in circulating cell-free DNA for early detection of esophageal cancer: a case-control study
Authors
Guibin Qiao
Weitao Zhuang
Bo Dong
Chengcheng Li
Jiayue Xu
Guoqiang Wang
Liang Xie
Zihao Zhou
Dan Tian
Gang Chen
Jiming Tang
Haiyu Zhou
Dongkun Zhang
Ruiqing Shi
Rixin Chen
Weiqi Nian
Yuzi Zhang
Jing Zhao
Xiaofang Wen
Yu Xu
Bingsi Li
Zhihong Zhang
Shangli Cai
Xiaosong Ben
Yu Qi
Publication date
01-12-2021
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2021
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-021-02109-y