Open Access

Value of circulating free DNA methylation in the diagnosis of esophageal cancer: A systematic review and meta‑analysis

  • Authors:
    • Chenxiao Ma
    • Yixian Wang
    • Zuopan Ding
    • Xingxing Liang
    • Meng Yuan
    • Xuan Liu
    • Qiong Li
    • Hongbo Lv
    • Na Qiao
    • Xin Liu
  • View Affiliations

  • Published online on: August 12, 2025     https://doi.org/10.3892/ol.2025.15225
  • Article Number: 478
  • Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liquid biopsy using circulating biomarkers has shown growing potential in cancer detection. The aim of the present study was to assess the application value of detecting the methylation level of circulating free (cf)DNA in the identification and diagnosis of esophageal cancer (EC), to lay a scientific foundation for its clinical application, and to provide statistically significant references for the detection process. A total of two researchers independently performed a comprehensive search of the PubMed, Web of Science, Cochrane Library, Embase and Scopus databases to identify all relevant studies on cfDNA in EC diagnosis as of July 25, 2024. A meta‑analysis was performed using MetaDiSc 1.4, Stata 16.0 and RevMan 5.4 software. Pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic rate (DOR) and area under the curve (AUC) values were determined. According to inclusion and exclusion criteria, a total of 14 studies with a total of 3,936 patients were included in the analysis. The pooled sensitivity and specificity, and PLR, NLR, DOR and AUC of cfDNA methylation in the diagnosis of EC were 0.83 [95% confidence interval (CI), 0.75‑0.89], 0.98 (95% CI, 0.95‑0.99), 34.57 (95% CI, 18.04‑63.44), 0.18 (95% CI, 0.12‑0.25), 197.25 (95% CI, 104.53‑372.20) and 0.98 (95% CI, 0.96‑0.99), respectively. In conclusion, cfDNA methylation demonstrates high diagnostic accuracy for early‑stage EC and may serve as a promising non‑invasive screening tool.

Introduction

As the global incidence and mortality rates of cancer continue to rise, it has emerged as a predominant public health concern, the leading cause of death worldwide and a major barrier to increasing life expectancy (1). Esophageal cancer (EC) is a prevalent malignant condition. It ranks among the most frequently diagnosed cancers globally, with the eighth highest incidence rate and the sixth highest mortality rate, with >500,000 new cases diagnosed annually, according to GLOBOCAN (2020) (1). A total of ~80% of new EC cases occur in developing countries, with ~60% of these cases diagnosed in China (2). Despite continuous advancements in diagnostic methods, surgical techniques and adjuvant chemoradiotherapy, most cases of esophageal squamous cell carcinoma (ESCC) are diagnosed at an advanced stage due to a lack of specific symptoms and preventive measures. The overall 5-year survival rate remains low, ranging from 15–50% (3,4). Therefore, effective treatment of EC greatly relies on the identification and prevention of the disease, and implementing screening and early detection methods is crucial for mitigating EC-related fatalities across all demographic groups.

Up to now, the detection of EC has continued to rely on non-molecular methods, which have limited specificity and sensitivity. The use of iodine staining (Lugol's solution) during endoscopic examination remains the gold standard for diagnosing irregularities in the esophageal mucosa (5). However, the widespread use of endoscopic screening in the population faces numerous challenges: Endoscopic targeted biopsy of the upper GI tract is unsuitable for mass screening of the general population due to its invasiveness, limited resources, low patient compliance and high cost, particularly when screening millions of eligible individuals in high-risk areas, such as China (6). Therefore, an urgent need exists to identify a more straightforward, simpler and precise approach for the early detection of EC.

Numerous studies have reported that DNA methylation, as a notable epigenetic alteration, serves a critical role in the progression of EC (7,8). However, traditional DNA methylation detection methods require tissue samples, which limits their clinical application (9). Liquid tumor biopsy, renowned for its procedural simplicity, high sensitivity, specificity and non-invasive or minimally invasive nature, facilitates dynamic monitoring of disease onset, progression and metastasis by detecting relevant tumor markers in the bloodstream, revolutionizing traditional methods of cancer diagnosis and treatment (10). Novel blood biomarkers, such as free DNA, free RNA, metabolites and proteins, have shown potential for the early detection of EC (1117).

Circulating free (cf)DNA is a type of extracellular DNA commonly found in the plasma or serum of patients with cancer, which has recently attracted the attention of researchers. It is released into the bloodstream by tumor cells (18) and shares the same genetic signatures and epigenetic modifications as tumor cells (19). This suggests that detecting cfDNA in serum could aid in the early diagnosis and treatment of cancer. Numerous studies have supported the potential of cfDNA as a biomarker for the early detection of cancer, making it a valuable analyte for liquid biopsy assessments (20,21), and offering a promising approach for non-invasive cancer detection. DNA methylation is widely recognized as one of the first molecular alterations occurring during cancer progression (22). Additionally, the biological and diagnostic applications of cell-free DNA have gained increasing popularity due to its minimally invasive, convenient and well-accepted characteristics (23). Abnormal methylated cfDNA detected in blood circulation appears to be a useful biomarker for human cancer (24). However, notable gaps remain in this field, with relatively few studies exploring the potential use of cfDNA methylation for the early detection of EC. Therefore, the present study aimed to thoroughly assess the practical use of cfDNA in diagnosing EC, evaluate the feasibility of cfDNA-based methylation as an early biomarker for EC, and provide a comprehensive reference for its clinical application.

Materials and methods

Search strategy and study selection

A total of two independent investigators performed a search across five databases, PubMed (https://pubmed.ncbi.nlm.nih.gov), Web of Science (https://www.webofscience.com), The Cochrane Library (https://www.cochranelibrary.com), Embase (https://www.embase.com) and Scopus (https://www.scopus.com), for all relevant articles published from the inception of each database until July 25, 2024. The search terms ‘esophageal neoplasms’, ‘esophageal cancers’, ‘cfDNA’, ‘circulating cell-free nucleic acid’ and ‘methylations’ were specified (Data S1). In addition, references in all relevant papers were assessed for potential research. The systematic review and meta-analysis were performed in strict accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analyses (25) guidelines and compliance with the established protocol, and is registered with the PROSPERO database (no. CRD42024580147; accessible at http://www.crd.york.ac.uk/prospero/).

Study selection

The inclusion criteria were as follows: i) Evaluation of the diagnostic value of cfDNA methylation in EC; ii) inclusion of malignant and benign EC; iii) patients had not undergone adjuvant therapy or received chemotherapy in the recent past, and had no record of other metastatic tumors prior to detection; iv) use of fresh serum samples for testing; v) endoscopy served as a diagnostic criterion for identifying EC; and vi) true positive, false positive, false negative and true negative values were calculated.

Moreover, the exclusion criteria were as follows: i) Non-English literature; ii) non-clinical research literature, including basic experiments, reviews, conference abstracts, laboratory notes and letters to journal editors; and iii) published data was not sufficient to construct a 2×2 table.

Data extraction

The literature was independently screened by two researchers, who extracted the data and subsequently cross-validated the results. Any studies with uncertain eligibility were addressed through consultation with an additional researcher. The data gathered encompassed the following: i) Fundamental characteristics of the studies, such as the principal investigator, date of publication, research methodology and the country of origin; ii) details pertaining to the demographic sample of the study, including the participant count and mean age; iii) techniques utilized for assessing methylation; and iv) the reported sensitivity and specificity metrics.

Quality assessment

To assess the quality of included studies, two reviewers independently performed quality assessments using the QUADAS-2 tool (version implemented: Original 2011 release with domain-specific signaling questions) (26), which was evidence-based and included patient selection, indicator detection, reference criteria, process and timing.

Statistical analysis

MetaDiSc 1.4 and Stata 16.0 software were employed for data analysis, and RevMan 5.4 software was used for quality assessment (2729). Initially, Meta-Disc was employed to assess the presence of threshold effects and heterogeneity. If the P-value associated with Spearman's correlation coefficient was >0.05, it was concluded that no threshold effect was present. Heterogeneity was evaluated using the Cochran-Q test, focusing on P-values and I2. P>0.05 and I2 ≤50% indicated no significant heterogeneity among the studies. If heterogeneity was detected, a subgroup analysis was performed to determine its source. The factors assessed in the subgroup analysis included the following: i) Ethnicity of the patients; ii) patient count; iii) mean age; and iv) techniques used for methylation detection. When a subgroup demonstrated P>0.05 and I2 ≤50%, covariates were considered as the origin of heterogeneity. The pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR) and diagnostic rate (DOR) were calculated using Stata 16.0, with their corresponding 95% confidence intervals (CI) represented by forest maps. The summary receiver operating characteristic curve was constructed, and the area under the curve (AUC) values were calculated. To assess publication bias, the Deeks funnel plot asymmetry test was employed. In all hypothesis tests, bilateral P<0.05 was considered to indicate a statistically significant difference.

Results

Study selection

In accordance with the research strategy, a total of 310 articles were sourced from online databases, including 2 from The Cochrane Library, 131 from Web of Science, 71 from Scopus, 99 from Embase and 7 from PubMed. These articles were subsequently screened according to the predefined inclusion and exclusion criteria. A total of 34 duplicates were removed, 113 reviews and conference proceedings were excluded, and 105 papers unrelated to the research topic were discarded based on their titles and abstracts. During the screening of the remaining 58 articles for full-text assessment, 44 papers were excluded: 12 were unrelated to cfDNA methylation in EC diagnosis, and 32 studies lacked sufficient data to construct a 2×2 table. Ultimately, a total of 14 studies were included in the analysis (12,3042). A visual representation of the literature screening process and its results is presented in Fig. 1.

Study characteristics

The meta-analysis incorporated 14 qualifying studies encompassing a collective patient count of 3,936, with an average participant number per study of 218. The articles were published between 2011 and 2024. Most of the articles were retrospective studies, and the majority of the patients were from Asia (8/14) and aged >60 years old on average. All included studies defined controls as allogeneic. Most studies detected cfDNA methylation used methylation-specific polymerase chain reaction (MSP). Table I lists the features of the included studies.

Table I.

Main features of the included studies.

Table I.

Main features of the included studies.

First author, yearCountryTPFPFNTNNo. of patientsMean age, yearsMethodsSample volume, mlControl sourceResearch design(Refs.)
Li et al, 2024China13443424741962EM-seq4HRetrospective(30)
Chen et al, 2024China783484169NRqMSP3HRetrospective(31)
Bian et al, 2024China29248426631,11655NR5HProspective(32)
Vavoulis et al, 2023UK602212967.5MSPNRHRetrospective(33)
Nicholson et al, 2023UK37699691,02165NR40HProspective(34)
Bian et al, 2023China1001918312449NRqMSPNRHRetrospective(35)
Qiao et al, 2021China653229418555MSPNRHRetrospective(12)
Liu et al, 2020USA47132071NREM-seq80HNR(36)
Wang et al, 2018China100031363MSP200HRetrospective(37)
Boldrin et al, 2020Europe1117322NRMSP1HRetrospective(38)
Zhai et al, 2012USA10002838NRNR0.1HRetrospective(39)
Wang et al, 2012China10101086026960MSPNRHNR(40)
Li et al, 2011China3708156060MSPNRHRetrospective(41)
Andolfo et al, 2011Europe33283275NRNR500HNR(42)

[i] TP, true positive; FP, false positive; FN, false negative; TN, true negative; MSP, methylation-specific polymerase chain reaction; qMSP, quantitative MSP; Control source, origin of baseline for comparison; H, heterogeneous (control samples from other individuals); EM-seq, enzymatic methyl-sequencing; NR, data not reported in the original studies.

Quality assessment and risk of bias

The quality assessment of the included studies, as per the QUADAS-2 scale, revealed that the collective quality of the literature was high (Fig. S1).

Accuracy of cfDNA methylation for EC

The pooled sensitivity and specificity were 0.83 (95% CI, 0.75–0.89) and 0.98 (95% CI, 0.95–0.99), respectively (Fig. 2), and the AUC was 0.98 (95% CI, 0.96–0.99; Fig. 3). Combined PLR and NLR were 34.57 (95% CI, 18.04–63.44) and 0.18 (95% CI, 0.12–0.26), respectively (Fig. S2), and combined DOR was 197.25 (95% CI, 104.53–372.20; Fig. S3).

Subgroup analysis

In the threshold analysis, a Spearman's correlation coefficient of 0.308 and a corresponding P-value of 0.284 were obtained, indicating the absence of a threshold effect. The I2 sensitivity and specificity results were 94.83 and 80.25%, respectively (Fig. 2), indicating the heterogeneity of sensitivity and specificity due to non-threshold effects. A subgroup analysis was then performed to assess the sources of heterogeneity. Subgroup analysis results indicated notable differences in diagnostic accuracy across subgroups based on sample size, patient age and cfDNA methylation test methods (Table II). Compared with subgroups with a sample size of <200 (DOR, 121.13; 95% CI, 50.83–288.63), those with a sample size of ≥200 exhibited markedly higher diagnostic accuracy (DOR, 172.17; 95% CI, 78.33–378.44). Moreover, compared with individuals aged ≤60 years (DOR, 12.21; 95% CI, 7.79–19.14), those aged >60 years exhibited notably higher diagnostic accuracy (DOR, 360.07; 95% CI, 174.49–743.02). Additionally, among the numerous methylation assessment methods, diagnostic accuracy using MSP (DOR, 66.21; 95% CI, 24.73–177.24) was markedly lower than that of other methods (DOR, 199.83; 95% CI, 83.56–477.85).

Table II.

Results of the subgroup analysis.

Table II.

Results of the subgroup analysis.

SensitivitySpecificityDOR



ClassificationnValue (95% CI)I2, %P-valueValue (95% CI)I2, %P-valueValue (95% CI)I2, %P-value
Ethnicity
  Caucasian60.83 (0.77–0.88)66.10.0100.99 (0.98–1.00)60.90.031136.64 (24.74–754.72)74.0<0.001
  Other80.78 (0.75–0.80)94.8<0.0010.95 (0.94–0.96)68.4<0.001114.33 (81.30–160.79)3.20.410
Number of patients
  <200100.71 (0.67–0.75)92.2<0.0010.97 (0.95–0.99)20.30.250121.13 (50.83–288.63)31.80.150
  ≥20040.85 (0.82–0.87)45.70.1400.97 (0.96–0.97)95.1<0.001172.17 (78.33–378.44)76.8<0.001
Mean age, years
  ≤6040.73 (0.70–0.77)97.0<0.0010.94 (0.92–0.96)74.20.01012.21 (7.79–19.14)0.00.991
  >6040.81 (0.75–0.85)33.90.2140.99 (0.99–1.00)0.00.502360.07 (174.49–743.02)0.00.400
Method
  MSP60.61 (0.56–0.66)87.8<0.0010.98 (0.95–0.99)36.70.16066.21 (24.73–177.24)7.20.374
  Other40.86 (0.82–0.89)80.8<0.0010.96 (0.94–0.97)58.70.062199.83 (83.56–477.85)50.70.112

[i] CI, confidence interval; DOR, diagnostic rate; MSP, methylation-specific polymerase chain reaction.

Sensitivity analysis and publication bias

A funnel plot was generated to assess publication bias, revealing a slope coefficient of P>0.05, and the plot appeared to be symmetrical, suggesting that no significant publication bias existed among the studies (Fig. S4).

Discussion

As a new diagnostic method for EC, numerous studies have reported that the detection of cfDNA methylation is an accurate and effective diagnostic method (19,20,24). Evidence suggests that cfDNA methylation achieves a sensitivity of 75–92% for early-stage detection, comparable with (and in certain cases exceeding) the 70–85% sensitivity of conventional endoscopic screening (43,44). Whilst endoscopy remains the gold standard for morphological assessment, its sensitivity can vary depending on operator experience and lesion characteristics (44). To date, no comprehensive review has yet been performed to assess the accuracy of the diagnosis, to the best of our knowledge. Therefore, the present systematic review and meta-analysis represents the first comprehensive attempt to evaluate the diagnostic efficacy of cfDNA methylation in EC. The findings indicate that the pooled sensitivity and specificity were 0.83 and 0.98, respectively, indicating high sensitivity and specificity for the differential diagnosis of EC, offering substantial support for clinical diagnosis of this disease.

cfDNA consists of small DNA fragments that are released into the bloodstream following cell death, and these fragments circulate throughout the system (45,46). In healthy individuals, cfDNA predominantly originates from blood-forming cells, though its composition can change under specific physiological or pathological conditions. DNA methylation, a widely investigated epigenetic modification, is characterized by the attachment of methyl groups to the carbon-5 position of cytosine, a process catalyzed by DNA methyltransferase enzymes, predominantly occurring at CpG dinucleotides (47).

An increasing body of evidence indicates that aberrant DNA methylation serves a notable role in the development and progression of cancer, primarily characterized by global hypomethylation, localized hypermethylation across several genomic regions (predominantly at CpG sites), and direct mutations in methylated cytosines (4850), which positions cfDNA as a potential biomarker for early cancer detection. Concurrently, research has demonstrated that the genetic data encapsulated within cfDNA can accurately identify tumor mutations and gene expression patterns (51). Moreover, the short half-life of cfDNA (ranging from 16 min to 2 h) enables dynamic monitoring of cancer progression (52,53). The methylation pattern of cfDNA is also reflective of the tissue or cell of origin, indicating that detecting aberrant DNA methylation patterns specific to tumors in the plasma of a patient could serve as a method for early cancer diagnosis and prognosis (54). Azad et al (55) reported an association between cfDNA levels in the blood of patients undergoing chemoradiotherapy for EC and tumor progression, metastasis and disease-specific survival rates. Similarly, Egyud et al (56) highlighted the ability of cfDNA to track treatment response and detect relapses in patients with esophageal adenocarcinoma (EAC). These findings align with the conclusions of the present review and provide the foundation for the present research.

Subgroup analyses based on sample size revealed that groups with larger sample sizes demonstrated improved diagnostic performance compared with those with smaller sample sizes. This could be attributed to the fact that elevated cfDNA levels are not exclusive to cancer but can also occur in conditions such as injury, heart attacks, strokes and chronic diseases such as diabetes. Several factors can influence cfDNA levels, including tumor size, location, vascularization and treatments such as surgery, chemotherapy, radiotherapy, and liver and kidney clearance mechanisms (57). Detection sensitivity is also affected by cfDNA molecule quantity, sequencing efficiency and the number of methylation markers. As sample size increases, the influence of these factors is diminished, enhancing diagnostic accuracy. In analyses of DNA methylation assay methods (using techniques such as enzymatic methyl-sequencing), diagnostic accuracy and sensitivity (other, 0.86; 95% CI, 0.82–0.89) and specificity (other, 0.96; 95% CI, 0.94–0.97) were markedly higher than those achieved with MSP methylation assay subgroups. It is recognized that cfDNA methylation identification methods generally fall into two categories: Bisulfite-based and bisulfite-free techniques. Bisulfite treatment involves converting unmethylated cytosines to uracil whilst leaving methylated cytosines unaffected, and the methylation status is determined using PCR amplification and sequencing (58). Bisulfite-free techniques refer to methodologies for detecting DNA methylation that do not rely on bisulfite conversion. Instead, they utilize alternative biochemical strategies to directly distinguish methylated cytosines (5mC/5hmC) from unmodified cytosines. Whole Genome Bisulfite Sequencing (WGBS) is considered the most comprehensive method for assessing DNA methylation, which constructs libraries and performs sequencing following the bisulfite conversion, yielding methylation data for every cytosine, encompassing areas with sparse CpG concentrations and non-CpG locations (CpA, CpT and CpC). Compared with WGBS, although WSP has a lower cost, there are few sites for study, and the design of primers or probes is complex, which may contribute to its lower sensitivity and specificity in certain subgroups (59).

Subgroup analysis by age revealed higher diagnostic accuracy in individuals aged >60 years compared with those aged <60 years. Despite a trend towards cancer incidence in younger individuals, EC remains more prevalent in middle-aged and older populations. The highest incidence peak occurs between 80–84 years of age (60). As age increases, the risk of precancerous lesions, multiple cancers and tumor heterogeneity rises, which may affect cfDNA concentration and methylation levels, influencing diagnostic accuracy.

In addition, EAC and ESCC are two distinct types of EC, with different origins and genetic characteristics (61). Although the present study was unable to perform subgroup analyses on using this variable due to insufficient data, preliminary judgments based on existing literature suggest that there are differences between EAC and ESCC. EAC demonstrates higher sensitivity (85–90%) using markers such as SRY-box transcription factor 17 and hyperplastic polyposis protein 1, whilst ESCC shows moderate sensitivity (75–82%) with markers such as cysteine dioxygenase type 1 or retinoic acid receptor β2 (62). These differences in sensitivity and the specific methylation markers highlight the distinct molecular pathways involved in the development of these two subtypes. However, future studies need to further explore these differences in larger sample sizes to provide more clinically meaningful guidance for subtype-specific applications.

Previous studies have reported that cfDNA methylation has strong diagnostic performance, with an early screening model achieving 74.7% sensitivity and 95.9% specificity (12,30). This outperforms traditional cytology and compares favorably with microRNA (miRNA)-based methods such as the combination of RUNX family transcription factor 3 methylation and carbohydrate antigen-19-9, which offer higher sensitivity in pancreatic cancer but lower specificity compared with cfDNA methylation (63). The superior stability of methylated DNA, compared with the susceptibility of miRNAs to degradation, may explain this advantage. Additionally, emerging technologies, such as machine learning-enhanced cfDNA methylation sequencing, further improve early detection accuracy (AUC, 0.91) and could markedly reduce cancer mortality (64). However, large-scale prospective trials are essential to validate these models across diverse populations and optimize detection thresholds.

Furthermore, despite its high diagnostic accuracy, the cost of cfDNA methylation testing remains a critical barrier to large-scale implementation. Technological advancements and economies of scale may help alleviate this challenge in the future. Furthermore, pre-analytical factors, such as blood collection methods, processing time and storage conditions, can affect test reproducibility. Standardized protocols are, therefore, crucial for clinical adoption. Whilst cfDNA methylation shows strong specificity, integrating it with other biomarkers, such as miRNAs or proteins, may offer a balanced approach for clinical implementation, particularly in resource-limited settings where cost-effectiveness is critical.

Moreover, there are certain limitations of the present study: i) EAC and ESCC were not classified in the study; ii) the impact of different stages of EC on diagnostic results was not taken into account; iii) due to the novelty of several detection techniques, there was no uniform cut-off value, which affected the accuracy of results; vi) the inclusion of English-only literature may have introduced selection bias; v) the findings of the present study were primarily derived from retrospective studies, which may have introduced selection bias and limits generalizability. Only two included studies employed prospective designs, underscoring the need for validation in large, multicenter prospective cohorts; and vi) most of the included studies involved patients from China, USA and UK, which may limit the generalizability of the findings to other populations.

In conclusion, the present study highlights the advancements in utilizing cfDNA for early EC diagnosis, substantiating the feasibility and efficacy of cfDNA methylation sequencing as a diagnostic modality. To the best of our knowledge, the present study is the first meta-analysis evaluating cfDNA methylation for EC diagnosis, demonstrating that the cfDNA methylation detection method has high sensitivity, specificity and AUC values. Moreover, analyzing serum cfDNA is a minimally invasive, rapid and efficient detection method and has potential diagnostic value for EC. However, diagnostic accuracy varies depending on several factors, such as the methylation sequencing methodology, cfDNA yield and cancer stage at detection. Therefore, additional prospective studies are required to validate the precision of cfDNA methylation detection across diverse parameters, facilitating optimization for clinical implementation. Future clinical trials should validate the clinical utility of cfDNA methylation for early EC detection, with potential integration into preventive healthcare paradigms. Such integration may notably improve early detection rates whilst reducing costs and enhancing patient safety.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

CM contributed to the conception and design of the study, data collection, data analysis, figure creation and drafting of the manuscript. YW and ZD confirm the authenticity of all raw data, contributed to the study design and participated in figure creation and critical revision of the manuscript. XingL, MY, XuanL, QL, HL and NQ contributed to the conception and design of the study and reviewed the manuscript. XinL contributed to the conception, design, review and revision of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI

2 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Rutegård M, Charonis K, Lu Y, Lagergren P, Lagergren J and Rouvelas I: Population-based esophageal cancer survival after resection without neoadjuvant therapy: An update. Surgery. 152:903–910. 2012. View Article : Google Scholar : PubMed/NCBI

4 

van Hagen P, Hulshof MC, van Lanschot JJ, Steyerberg EW, van Berge Henegouwen MI, Wijnhoven BP, Richel DJ, Nieuwenhuijzen GA, Hospers GA, Bonenkamp JJ, et al: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 366:2074–2084. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Liang H, Fan JH and Qiao YL: Epidemiology, etiology, and prevention of esophageal squamous cell carcinoma in China. Cancer Biol Med. 14:33–41. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Li H, Ding C, Zeng H, Zheng R, Cao M, Ren J, Shi J, Sun D, He S, Yang Z, et al: Improved esophageal squamous cell carcinoma screening effectiveness by risk-stratified endoscopic screening: Evidence from high-risk areas in China. Cancer Commun (Lond). 41:715–725. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Li P, Liu X, Dong ZM and Ling ZQ: Epigenetic silencing of HIC1 promotes epithelial-mesenchymal transition and drives progression in esophageal squamous cell carcinoma. Oncotarget. 6:38151–38165. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Xu T, Ding H, Chen J, Lei J, Zhao M, Ji B, Chen Y, Qin S and Gao Q: Research progress of DNA methylation in endometrial cancer. Biomolecules. 12:9382022. View Article : Google Scholar : PubMed/NCBI

9 

Zhang J, Sheng H, Hu C, Li F, Cai B and Ma Y, Wang Y and Ma Y: Effects of DNA methylation on gene expression and phenotypic traits in cattle: A review. Int J Mol Sci. 24:118822023. View Article : Google Scholar : PubMed/NCBI

10 

Peneder P, Stütz AM, Surdez D, Krumbholz M, Semper S, Chicard M, Sheffield NC, Pierron G, Lapouble E, Tötzl M, et al: Multimodal analysis of cell-free DNA whole-genome sequencing for pediatric cancers with low mutational burden. Nat Commun. 12:32302021. View Article : Google Scholar : PubMed/NCBI

11 

Miyoshi J, Zhu Z, Luo A, Toden S, Zhou X, Izumi D, Kanda M, Takayama T, Parker IM, Wang M, et al: A microRNA-based liquid biopsy signature for the early detection of esophageal squamous cell carcinoma: A retrospective, prospective and multicenter study. Mol Cancer. 21:442022. View Article : Google Scholar : PubMed/NCBI

12 

Qiao G, Zhuang W, Dong B, Li C, Xu J, Wang G, Xie L, Zhou Z, Tian D, Chen G, et al: Discovery and validation of methylation signatures in circulating cell-free DNA for early detection of esophageal cancer: A case-control study. BMC Med. 19:2432021. View Article : Google Scholar : PubMed/NCBI

13 

Qin Y, Wu CW, Taylor WR, Sawas T, Burger KN, Mahoney DW, Sun Z, Yab TC, Lidgard GP, Allawi HT, et al: Discovery, validation, and application of novel methylated DNA markers for detection of esophageal cancer in plasma. Clin Cancer Res. 25:7396–7404. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Xue Y, Wang K, Jiang Y, Dai Y, Liu X, Pei B, Li H, Xu H and Zhao G: An ultrasensitive and multiplexed miRNA one-step real time RT-qPCR detection system and its application in esophageal cancer serum. Biosens Bioelectron. 247:1159272024. View Article : Google Scholar : PubMed/NCBI

15 

Lu D, Wu X, Wu W, Wu S, Li H, Zhang Y, Yan X, Zhai J, Dong X, Feng S, et al: Plasma cell-free DNA 5-hydroxymethylcytosine and whole-genome sequencing signatures for early detection of esophageal cancer. Cell Death Dis. 14:8432023. View Article : Google Scholar : PubMed/NCBI

16 

Yang X, Suo C, Zhang T, Yin X, Man J, Yuan Z, Yu J, Jin L, Chen X, Lu M and Ye W: Targeted proteomics-derived biomarker profile develops a multi-protein classifier in liquid biopsies for early detection of esophageal squamous cell carcinoma from a population-based case-control study. Biomark Res. 9:122021. View Article : Google Scholar : PubMed/NCBI

17 

Lv J, Wang J, Shen X, Liu J, Zhao D, Wei M, Li X, Fan B, Sun Y, Xue F, et al: A serum metabolomics analysis reveals a panel of screening metabolic biomarkers for esophageal squamous cell carcinoma. Clin Transl Med. 11:e4192021. View Article : Google Scholar : PubMed/NCBI

18 

Chan KC, Jiang P, Chan CW, Sun K, Wong J, Hui EP, Chan SL, Chan WC, Hui DS, Ng SS, et al: Noninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencing. Proc Natl Acad Sci USA. 110:18761–18768. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Hattori N and Ushijima T: Compendium of aberrant DNA methylation and histone modifications in cancer. Biochem Biophys Res Commun. 455:3–9. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Jiang M, Zhou J, Xie X, Huang Z, Liu R and Lv Y: Single nanoparticle counting-based liquid biopsy for cancer diagnosis. Anal Chem. 94:15433–15439. 2022. View Article : Google Scholar : PubMed/NCBI

21 

Llueca A, Canete-Mota S, Jaureguí A, Barneo M, Ibañez MV, Neef A, Ochoa E, Tomas-Perez S, Mari-Alexandre J, Gilabert-Estelles J, et al: The impact of liquid biopsy in advanced ovarian cancer care. Diagnostics (Basel). 14:18682024. View Article : Google Scholar : PubMed/NCBI

22 

Weinberg DN, Rosenbaum P, Chen X, Barrows D, Horth C, Marunde MR, Popova IK, Gillespie ZB, Keogh MC, Lu C, et al: Two competing mechanisms of DNMT3A recruitment regulate the dynamics of de novo DNA methylation at PRC1-targeted CpG islands. Nat Genet. 53:794–800. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Nie K, Jia Y and Zhang X: Cell-free circulating tumor DNA in plasma/serum of non-small cell lung cancer. Tumour Biol. 36:7–19. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Li X, Zhou F, Jiang C, Wang Y, Lu Y, Yang F, Wang N, Yang H, Zheng Y and Zhang J: Identification of a DNA methylome profile of esophageal squamous cell carcinoma and potential plasma epigenetic biomarkers for early diagnosis. PLoS One. 9:e1031622014. View Article : Google Scholar : PubMed/NCBI

25 

Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al: The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 372:n712021. View Article : Google Scholar : PubMed/NCBI

26 

Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, Leeflang MM, Sterne JA and Bossuyt PM; QUADAS-2 Group, : QUADAS-2: A revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 155:529–536. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Zamora J, Abraira V, Muriel A, Khan K and Coomarasamy A: Meta-DiSc: A software for meta-analysis of test accuracy data. BMC Med Res Methodol. 6:312006. View Article : Google Scholar : PubMed/NCBI

28 

StataCorp. Stata Statistical Software, . Release 16. College Station, TX: StataCorp LLC; 2019, https://www.stata.comJuly 25–2024

29 

Review Manager (RevMan) [Computer program], . Version 5.4. The Cochrane Collaboration. 2020.Available from. https://training.cochrane.org/online-learning-materials/revman

30 

Li Y, Liu B, Zhou X, Yang H, Han T, Hong Y, Wang C, Huang M, Yan S, Li S, et al: Genome-Scale multimodal analysis of cell-free DNA whole-methylome sequencing for noninvasive esophageal cancer detection. JCO Precis Oncol. 8:e24001112024. View Article : Google Scholar : PubMed/NCBI

31 

Chen X, Li M, Li Y, Aiolfi A, Bonavina L, Lerut T, Wu X and Zhang Q: Combining non-invasive liquid biopsy and a methylation analysis to assess surgical risk for early esophageal cancer. Transl Cancer Res. 13:3075–3089. 2024. View Article : Google Scholar : PubMed/NCBI

32 

Bian Y, Gao Y, Lin H, Sun C, Wang W, Sun S, Li X, Feng Z, Ren J, Chen H, et al: Non-invasive diagnosis of esophageal cancer by a simplified circulating cell-free DNA methylation assay targeting OTOP2 and KCNA3: A double-blinded, multicenter, prospective study. J Hematol Oncol. 17:472024. View Article : Google Scholar : PubMed/NCBI

33 

Vavoulis D, Cutts A, Thota N, Brown J, Sugar R, Rueda A, Ardalan A, Matos Santo F, et al: Multimodal cell-free DNA whole-genome analysis combined with TET-Assisted Pyridine Borane Sequencing is sensitive and reveals specific cancer signals. MedRxiv. DOI:. https://doi.org/10.1101/2023.09.29.23296336

34 

Nicholson BD, Oke J, Virdee PS, Harris DA, O'Doherty C, Park JE, Hamady Z, Sehgal V, Millar A, Medley L, et al: Multi-cancer early detection test in symptomatic patients referred for cancer investigation in England and Wales (SYMPLIFY): a large-scale, observational cohort study. Lancet Oncol. 24:733–743. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Bian Y, Gao Y, Lu C, Tian B, Xin L, Lin H, Zhang Y, Zhang X, Zhou S, Wan K, et al: Genome-wide methylation profiling identified methylated KCNA3 and OTOP2 as promising diagnostic markers for esophageal squamous cell carcinoma. Chin Med J (Engl). 137:1724–1735. 2024. View Article : Google Scholar : PubMed/NCBI

36 

Liu MC, Oxnard GR, Klein EA, Swanton C and Seiden MV; CCGA Consortium, : Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol. 31:745–759. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Wang HQ, Yang CY, Wang SY, Wang T, Han JL, Wei K, Liu FC, Xu JD, Peng XZ and Wang JM: Cell-free plasma hypermethylated CASZ1, CDH13 and ING2 are promising biomarkers of esophageal cancer. J Biomed Res. 32:424–433. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Boldrin E, Curtarello M, Dallan M, Alfieri R, Realdon S, Fassan M and Saggioro D: Detection of LINE-1 hypomethylation in cfDNA of esophageal adenocarcinoma patients. Int J Mol Sci. 21:15472020. View Article : Google Scholar : PubMed/NCBI

39 

Zhai R, Zhao Y, Su L, Cassidy L, Liu G and Christiani DC: Genome-wide DNA methylation profiling of cell-free serum DNA in esophageal adenocarcinoma and Barrett esophagus. Neoplasia. 14:29–33. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Wang CC, Mao WM and Ling ZQ: DNA methylation status of RARβ2 and p16(INK4α) in peripheral blood and tumor tissue in patients with esophageal squamous cell carcinoma. Zhonghua Zhong Liu Za Zhi. 34:441–445. 2012.(In Chinese). PubMed/NCBI

41 

Li B, Wang B, Niu LJ, Jiang L and Qiu CC: Hypermethylation of multiple tumor-related genes associated with DNMT3b up-regulation served as a biomarker for early diagnosis of esophageal squamous cell carcinoma. Epigenetics. 6:307–316. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Andolfo I, Petrosino G, Vecchione L, De Antonellis P, Capasso M, Montanaro D, Gemei M, Troncone G, Iolascon A, Orditura M, et al: Detection of erbB2 copy number variations in plasma of patients with esophageal carcinoma. BMC Cancer. 11:1262011. View Article : Google Scholar : PubMed/NCBI

43 

Constantin N, Sina AA, Korbie D and Trau M: Opportunities for early cancer detection: The rise of ctDNA methylation-based pan-cancer screening technologies. Epigenomes. 6:62022. View Article : Google Scholar : PubMed/NCBI

44 

Nikanjam M, Kato S and Kurzrock R: Liquid biopsy: Current technology and clinical applications. J Hematol Oncol. 15:1312022. View Article : Google Scholar : PubMed/NCBI

45 

Lo YM, Chan KC, Sun H, Chen EZ, Jiang P, Lun FM, Zheng YW, Leung TY, Lau TK, Cantor CR and Chiu RW: Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med. 2:61ra912010. View Article : Google Scholar : PubMed/NCBI

46 

Heitzer E, Auinger L and Speicher MR: Cell-Free DNA and apoptosis: How dead cells inform about the living. Trends Mol Med. 26:519–528. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Jones PA: Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 13:484–492. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Esteller M: Epigenetics in cancer. N Engl J Med. 358:1148–1159. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Baylin SB and Jones PA: A decade of exploring the cancer epigenome-biological and translational implications. Nat Rev Cancer. 11:726–734. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Dor Y and Cedar H: Principles of DNA methylation and their implications for biology and medicine. Lancet. 392:777–786. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Esfahani MS, Hamilton EG, Mehrmohamadi M, Nabet BY, Alig SK, King DA, Steen CB, Macaulay CW, Schultz A, Nesselbush MC, et al: Inferring gene expression from cell-free DNA fragmentation profiles. Nat Biotechnol. 40:585–597. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Lo YM, Corbetta N, Chamberlain PF, Rai V, Sargent IL, Redman CW and Wainscoat JS: Presence of fetal DNA in maternal plasma and serum. Lancet. 350:485–487. 1997. View Article : Google Scholar : PubMed/NCBI

53 

Yu SC, Lee SW, Jiang P, Leung TY, Chan KC, Chiu RW and Lo YM: High-resolution profiling of fetal DNA clearance from maternal plasma by massively parallel sequencing. Clin Chem. 59:1228–1237. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Feng H, Jin P and Wu H: Disease prediction by cell-free DNA methylation. Brief Bioinform. 20:585–597. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Azad TD, Chaudhuri AA, Fang P, Qiao Y, Esfahani MS, Chabon JJ, Hamilton EG, Yang YD, Lovejoy A, Newman AM, et al: Circulating tumor DNA analysis for detection of minimal residual disease after chemoradiotherapy for localized esophageal cancer. Gastroenterology. 158:494–505.e6. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Egyud M, Tejani M, Pennathur A, Luketich J, Sridhar P, Yamada E, Ståhlberg A, Filges S, Krzyzanowski P, Jackson J, et al: Detection of circulating tumor DNA in plasma: A potential biomarker for esophageal adenocarcinoma. Ann Thorac Surg. 108:343–349. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Fleischhacker M and Schmidt B: Circulating nucleic acids (CNAs) and cancer-a survey. Biochim Biophys Acta. 1775:181–232. 2007.PubMed/NCBI

58 

Frommer M, McDonald LE, Millar DS, Collis CM, Watt F, Grigg GW, Molloy PL and Paul CL: A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci USA. 89:1827–1831. 1992. View Article : Google Scholar : PubMed/NCBI

59 

Huang J and Wang L: Cell-Free DNA methylation profiling analysis-technologies and bioinformatics. Cancers (Basel). 11:17412019. View Article : Google Scholar : PubMed/NCBI

60 

Zhu H, Ma X, Ye T, Wang H, Wang Z, Liu Q and Zhao K: Esophageal cancer in China: Practice and research in the new era. Int J Cancer. 152:1741–1751. 2023. View Article : Google Scholar : PubMed/NCBI

61 

Lu T, Chen D, Wang Y, Sun X, Li S, Miao S, Wo Y, Dong Y, Leng X, Du W and Jiao W: Identification of DNA methylation-driven genes in esophageal squamous cell carcinoma: a study based on The Cancer Genome Atlas. Cancer Cell Int. 19:522019. View Article : Google Scholar : PubMed/NCBI

62 

Xu Y, Wang Z, Pei B, Wang J, Xue Y and Zhao G: DNA methylation markers in esophageal cancer. Front Genet. 15:13541952024. View Article : Google Scholar : PubMed/NCBI

63 

Bixby B, Vrba L, Lenka J, Oshiro M, Watts GS, Hughes T, Erickson H, Chopra M, Knepler JL, Knox KS, et al: Cell-Free DNA methylation analysis as a marker of malignancy in pleural fluid. Sci Rep. Feb 5–2023.(Epub ahead of print).

64 

Zhou X, Cheng Z, Dong M, Liu Q, Yang W, Liu M, Tian J and Cheng W: Tumor fractions deciphered from circulating cell-free DNA methylation for cancer early diagnosis. Nat Commun. 13:76942022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2025
Volume 30 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma C, Wang Y, Ding Z, Liang X, Yuan M, Liu X, Li Q, Lv H, Qiao N, Liu X, Liu X, et al: Value of circulating free DNA methylation in the diagnosis of esophageal cancer: A systematic review and meta‑analysis. Oncol Lett 30: 478, 2025.
APA
Ma, C., Wang, Y., Ding, Z., Liang, X., Yuan, M., Liu, X. ... Liu, X. (2025). Value of circulating free DNA methylation in the diagnosis of esophageal cancer: A systematic review and meta‑analysis. Oncology Letters, 30, 478. https://doi.org/10.3892/ol.2025.15225
MLA
Ma, C., Wang, Y., Ding, Z., Liang, X., Yuan, M., Liu, X., Li, Q., Lv, H., Qiao, N., Liu, X."Value of circulating free DNA methylation in the diagnosis of esophageal cancer: A systematic review and meta‑analysis". Oncology Letters 30.4 (2025): 478.
Chicago
Ma, C., Wang, Y., Ding, Z., Liang, X., Yuan, M., Liu, X., Li, Q., Lv, H., Qiao, N., Liu, X."Value of circulating free DNA methylation in the diagnosis of esophageal cancer: A systematic review and meta‑analysis". Oncology Letters 30, no. 4 (2025): 478. https://doi.org/10.3892/ol.2025.15225