Next Article in Journal
Impact of Chemotherapy Dose Intensity on Pathological Complete Response in Pembrolizumab-Treated Early Triple-Negative Breast Cancer: A Real-World Multicenter Analysis
Previous Article in Journal
Sugammadex Versus Neostigmine in Return to Intended Oncological Therapy After Gastrointestinal Cancer Surgery: A Retrospective Study
Previous Article in Special Issue
Oral Viral DNA Profiling in Obesity, Adenomatous Polyposis, and Colorectal Cancer Identifies Human β-Papillomavirus Types as Potentially Sex-Related and Modifiable Cancer Risk Indicators
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Blood-Based Surveillance Biomarkers for Gastroesophageal Cancers

1
Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA
2
Department of Molecular, Cellular & Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(21), 3552; https://doi.org/10.3390/cancers17213552
Submission received: 1 October 2025 / Revised: 26 October 2025 / Accepted: 31 October 2025 / Published: 2 November 2025

Simple Summary

Gastroesophageal cancers are often diagnosed late and frequently recur. Current surveillance methods are invasive and sometimes inadequate. This review examines “liquid biopsies”—blood tests that detect cancer markers like circulating tumor DNA (ctDNA), methylated DNA, and other tumor-derived substances. These tests offer a minimally invasive way to monitor patients. While ctDNA and methylated DNA show particular promise for earlier recurrence detection compared to imaging, most blood-based biomarkers are still investigational. The integration of multi-analyte assays, AI, and ongoing clinical trials suggests that liquid biopsies will soon become a transformative tool for improved surveillance and personalized care in gastroesophageal cancers.

Abstract

Gastroesophageal cancers including esophageal and gastric cancer remain major causes of global cancer mortality, primarily due to late diagnosis and high recurrence rates after curative treatment. Current surveillance methods, such as endoscopy and imaging, are invasive, costly, and often inadequate for detection. Blood-based biomarkers (“liquid biopsies”) offer a minimally invasive alternative capable of real-time tumor monitoring. In this review, we summarize recent advances across all major classes of blood-derived biomarkers: circulating tumor DNA (ctDNA), methylated DNA, cell-free RNAs (microRNAs, lncRNAs, circRNAs), circulating proteins, autoantibodies, circulating tumor cells, extracellular vesicles, and metabolites. Reviewing the existing literature on gastroesophageal cancers, we highlight current evidence, validation phases, performance metrics, and limitations. Special attention is given to clinical trial evidence, including ctDNA monitoring studies, that demonstrated earlier recurrence detection compared to imaging. While blood-based biomarker analysis has not yet supplanted endoscopy as standard of care in gastroesophageal cancer surveillance, the convergence of multi-analyte assays, AI, and clinical validation trials positions liquid biopsy as a transformative tool in the surveillance of gastroesophageal cancers.

1. Introduction

Gastric and esophageal cancers are a significant global health burden, ranking among the top five cancers for diagnosis and mortality in 2022 [1]. Esophageal cancer also poses a substantial threat, with an estimated 511,054 new cases and 445,391 deaths globally in 2022 [2]. Their lethality stems from non-specific symptoms (nausea, pain, early satiety) that delay diagnosis, often resulting in peritoneal metastasis at presentation. This leads to poor prognoses, with 5-year survival for esophageal cancer under 25% and advanced gastric cancer survival around 12 months [3,4,5].
Current treatments include surgery (esophagectomy, gastrectomy), chemotherapy (neoadjuvant, adjuvant, palliative, e.g., FOLFOX, cisplatin, FLOT, CAPOX), radiation, and targeted therapies (trastuzumab for HER2+, immune checkpoint inhibitors) [6,7]. However, recurrence rates are high (40–60%), indicating a critical need for more effective surveillance strategies [8] highlights the need for more effective surveillance strategies.
There is an unmet need for minimally invasive surveillance strategies that can detect cancer recurrence earlier and more readily than conventional methods. Circulating biomarkers originate through tumor apoptosis, necrosis, and active secretion of nucleic acids and vesicles into the bloodstream. These fragments reflect tumor burden, genetic heterogeneity, and host–immune interactions, providing a minimally invasive window into disease dynamics. Blood-based biomarkers measure tumor-derived or tumor-associated factors circulating in peripheral blood, offering a promising approach for such surveillance. These “liquid biopsies” offer potential advantages, including safety, repeatability, cost-effectiveness, and patient acceptability, which enable real-time tumor monitoring with higher compliance. Earlier detection of recurrence through blood-based surveillance could enable more timely intervention with salvage therapies, leading to improved patient outcomes and potentially personalized treatment approaches. In this review, we provide an overview of the major classes of blood assays and biomarkers under investigation, detail current and emerging biomarkers for each gastroesophageal (GE) cancers subtype, esophageal squamous cell carcinoma (ESCC) vs. adenocarcinoma and gastric adenocarcinoma (EAC), and discuss their mechanisms, performance metrics, regulatory status, and limitations. Diagnostic biomarkers enable early detection, prognostic biomarkers stratify outcomes or therapeutic response, and surveillance biomarkers monitor minimal residual disease (MRD) and recurrence. This review focuses on the latter, emphasizing translational and clinical readiness. Relevant publications were identified through targeted searches of PubMed, Embase, and ClinicalTrials.gov (2010–2025) using combinations of ‘gastroesophageal cancers’, ‘liquid biopsy’, ‘ctDNA’, and ‘surveillance’.

2. Overview of Blood-Based Biomarker Assays and Classes

Multiple classes of blood-based biomarkers are being explored for cancer detection and surveillance. Each class offers distinct biological insights and technical considerations:
After curative-intent treatment for GE cancer, in both gastric and esophageal cancers, ctDNA is utilized as a prognostic tool to monitor for MRD or potential relapse [9]. Postsurgical ctDNA positivity is a strong indicator of recurrence, and longitudinal monitoring with highly sensitive, personalized assays can enable earlier detection of relapse compared to standard imaging methods [10]. This allows for better risk stratification: patients with ctDNA-positive results may benefit from more intensive surveillance or consideration for adjuvant therapy, while a negative result could potentially lead to de-escalation of treatment and prevent unnecessary toxicity [10]. Although several ctDNA tests are commercially available and some are FDA-approved for advanced cancer monitoring, their use for surveillance in GE cancer remains in the investigational stage [9,11].
Circulating cell-free RNA (cfRNA), which includes microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), is being investigated as a biomarker for cancer surveillance [12]. These RNA molecules are released by tumor cells and are appealing for their stability in the blood [12,13]. Since cfRNA markers often reflect oncogenic or tumor-suppressive pathways, they offer promise for monitoring cancer status, including the detection of minimal residual disease or recurrence [14]. However, despite promising research and some markers entering validation phases, no cfRNA-based blood testing is yet in routine clinical use for GE malignancies. Challenges include ensuring assay specificity, and ongoing research is necessary to fully validate its use in clinical practice [12,15].
Despite their decade-long use, soluble protein tumor markers exhibit suboptimal performance in surveillance for GE cancers due to several factors. Their limited sensitivity in detecting early recurrence, coupled with high false-positive rates due to inflammatory conditions, benign diseases, and lifestyle factors, compromises their reliability. The genetic limitation affecting CA19-9 production in Lewis-negative individuals further contributes to false negatives. The variable prognostic value and lack of standardized interpretation add to the challenges. As such, relying solely on soluble protein tumor markers for effective surveillance is insufficient, prompting research into combining them with other biomarkers to improve sensitivity and specificity [16,17]. Common examples include carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9), and carbohydrate antigen 72-4 (CA72-4), among others [18]. Immunoassays detect these markers but generally exhibit poor sensitivity for early-stage disease [16]. Their moderate specificity can be compromised by benign conditions, resulting in false positives [16]. As a result, research is now focusing on combining protein markers with other modalities to improve detection sensitivity [16,17].
The immune system’s early recognition of tumor-associated antigens (TAAs) can trigger an autoantibody response that can be measured for cancer surveillance [19]. These autoantibodies are highly specific for cancer and stable in serum, with some being detectable months or years before a clinical cancer diagnosis [19,20]. Single autoantibodies, such as those against p53 or NY-ESO-1, are often found in only a minority of patients, so panels of multiple autoantibodies are used to increase sensitivity [20,21]. This approach is challenged by patient-specific antibody repertoires and the need to differentiate between cancer-induced antibodies and those from autoimmune conditions [20]. While multiplex autoantibody tests exist for other cancers, such as the Early CDT platform for lung cancer, none are currently standard for surveillance in GE cancers [22,23]. Autoantibody panels are currently under evaluation in clinical cohorts for esophageal and gastric cancers.
Intact cancer cells that have detached from a primary or metastatic tumor and entered the bloodstream are known as circulating tumor cells (CTCs) [24]. Their presence generally correlates with advanced disease and a worse prognosis [24]. For surveillance purposes, a persistent or rising CTC count after surgery can indicate residual disease and allow for real-time genotyping of the tumor [24,25]. However, in GE cancers, CTC assays face challenges due to technical limitations and low sensitivity, particularly in early-stage or non-metastatic disease [24,26,27]. While emerging microfluidic and size-based capture methods are improving detection, CTC-based tests are not yet standard for GE cancer surveillance, though research is ongoing [25,26,27].
Extracellular vesicles (EVs), including exosomes, are actively secreted by tumor cells and contain a payload of proteins, DNA, and RNA that reflect their cell of origin [28]. The contents of these vesicles are protected from degradation, making them stable in blood and useful as biomarkers for surveillance [29]. Tumor-derived exosomes, for instance, have been shown to contain characteristic miRNAs and proteins that can serve as biomarkers in GE cancers [30]. Exosomal DNA with cancer-specific mutations and various miRNAs have been studied for their diagnostic potential, and EVs in general contribute to intercellular communication [29,30]. However, challenges persist with the specialized equipment required for isolation and characterization, as well as the lack of assay standardization [29]. While EV-based diagnostics are not yet clinically utilized for GE malignancies, research is ongoing, with the potential for these assays to one day complement other liquid biopsy methods [29].
Tumor-specific methylation changes in cell-free DNA (cfDNA) serve as highly promising biomarkers for cancer surveillance [31,32]. Aberrant promoter methylation can act as a specific cancer signal in the blood, reflecting early events in carcinogenesis and holding promise for detecting premalignant or early changes [32,33,34]. cfDNA methylation markers are under active investigation for GE cancers [33,35]. For gastric cancer, candidate markers include P16, RASSF1A, RPRM, and RUNX3, among others, with some studies demonstrating strong performance [33,36]. In esophageal cancer, various methylated DNA markers, including SFRP1, TAC1, and P16, have been detected in circulation [37].
Tumors cause systemic metabolic changes detectable in blood through metabolomics [38]. Studies have shown altered metabolite levels in gastric cancer that partially normalize after tumor removal, suggesting their potential for monitoring biochemical recurrence [39].
Based on the current landscape of liquid biopsy assays, ctDNA and tumor-specific methylation changes in cfDNA appear promising for surveillance in GE cancers. ctDNA is already used as a prognostic tool and can enable earlier relapse detection, while cfDNA methylation markers show promise for detecting early cancer events. However, analytes like soluble protein tumor markers, circulating tumor cells (CTCs), circulating cell-free RNA (cfRNA), autoantibodies, extracellular vesicles (EVs) and metabolomics require further refinement of detection and validation before they can be routinely used for early detection and surveillance in GE cancers.
To better inform surveillance, blood-based biomarker panels are increasingly recognized as a non-invasive and comprehensive method for monitoring GE cancers. Rather than relying on a single marker, which offers limited sensitivity, combining various biomarker types such as ctDNA mutations/methylation, cancer-associated miRNAs, and protein markers allows for the capture of multiple tumor signatures. This multi-modal approach improves overall sensitivity and specificity by providing a more complete picture of tumor activity. This blood-based liquid biopsy approach offers a significant advantage for surveillance, as samples can be collected serially and less invasively than traditional biopsies, offering the potential for more frequent monitoring. In contrast, traditional surveillance methods like imaging often involve radiation exposure, logistical difficulties, and significant costs, which limit their practicality for frequent use. These limitations, combined with the inherent challenges in achieving high accuracy and precision in imaging-based tumor monitoring, can lengthen the lead time for recurrence detection. Addressing these limitations, regular, non-invasive blood-based testing enables timely and accurate monitoring of high-risk individuals and postoperative patients, offering the potential for earlier detection of recurrence or progression. Additionally, by analyzing multiple circulating components, these assays provide a dynamic assessment of a tumor’s molecular landscape over time, which could guide personalized treatment strategies and potentially improve outcomes. Comparing across biomarker modalities reveals complementary strengths. ctDNA provides high specificity for molecular relapse but limited sensitivity in early disease, whereas cfRNA and EVs capture active tumor biology. Protein and metabolite markers remain inexpensive but lack tumor specificity. Analytical limitations include cfRNA degradation, exosome isolation variability, and CHIP-related false positives. AI-assisted multi-omics pipelines integrating methylation, proteomic, and fragmentomic data represent promising directions toward clinically deployable, high-accuracy surveillance tools [40,41]. Figure 1 provides a visual overview of the different types of liquid biopsy assays used in the surveillance of GE cancers. It highlights the specific biomarkers detected by each assay type and their clinical applications in esophageal and gastric cancers. Table 1 summarizes key blood-based biomarkers evaluated across GE cancers. Among ongoing efforts, studies integrating ctDNA methylation and fragmentomics (e.g., GUIDE and GutSeer) appear particularly promising because they combine genomic and epigenetic signals with high reported sensitivity and specificity. Reported sensitivities and specificities vary widely across studies depending on stage (localized vs. metastatic), assay platform, and study design. Unless otherwise stated, metrics represent diagnostic rather than surveillance contexts. Where available, 95% CIs have been added. Together, the figure and the table provide a comprehensive overview of the current landscape of liquid biopsy biomarkers in GE cancer surveillance.
While numerous studies report promising sensitivity and specificity for blood-based biomarkers, several others have shown limited or inconsistent diagnostic utility. For instance, meta-analyses of CEA and CA19-9 in gastric cancer demonstrate substantial heterogeneity, with pooled sensitivities often below 40% due to variable assay thresholds, patient stage distribution, and ethnic differences in Lewis antigen expression. These discrepancies highlight the need for harmonized assays and prospective validation before clinical implementation [42,43]. Ethnic and environmental factors may significantly influence biomarker profiles. Variations in diet, Helicobacter pylori prevalence, and genetic polymorphisms (e.g., Lewis antigen phenotype affecting CA19-9) contribute to population-specific performance differences [42]. Consequently, biomarker panels validated in East Asian cohorts may not directly translate to Western populations, underscoring the need for global multicenter validation.
Table 1. Comparative overview of blood-based biomarker classes in gastroesophageal cancer surveillance.
Table 1. Comparative overview of blood-based biomarker classes in gastroesophageal cancer surveillance.
Biomarker (Assay)Cancer TypeClinical ContextSensitivity/SpecificityKey Analytical LimitationValidation PhaseSource(s)
CEA (Carcinoembryonic antigen, ELISA) Class: Protein (Oncofetal antigen)Esophageal adenocarcinoma; Gastric adenocarcinomaSurveillance for recurrence in advanced disease (also diagnostic adjunct)EC (detection):
Se = 27.5% (18.9–35.2%)
Sp = 95.4% (94.1–96.8%)
Esophageal (recurrence):
Se = 54.7% (40.9–67.8%)
Sp = 90.0% (73.5–97.9%)
GC (detection):
Se = 20.1% (18.3–22.1%),
Sp = 94.7% (93.6–95.7%)
Gastric (recurrence):
Se = 73.0% (68.8–77.2%),
Sp = 59.0% (56.3–61.7%)
Low sensitivity and specificity in early-stage cancerEsophagus: Phase 2
Gastric: Phase 3
[44,45,46,47,48,49]
CA19-9 (Carbohydrate Antigen 19-9): ELISA; Class: Protein (glycan antigen)Gastric adenocarcinoma (subset); Advanced disease monitoring (especially in pancreatobiliary-type or intestinal-type tumors)GC (detection):
Se = 21.4% (19.3–23.0%)
Sp = 96.2% (95.2–97.1%)
Gastric (Recurrence):
Se = 24.1% (10.3–43.5%),
Sp = 93.3% (87.3–97.1%)
Poor sensitivity, low specificity, false-negative results in some patient populations, and a lack of standardized cut-off values further hinders its reliable usePhase 3[48,49]
CA72-4 (Carbohydrate Antigen 72-4): ELISA; Class: Protein (glycoprotein antigen)Gastric adenocarcinomaDiagnostic adjunct; recurrence monitoringDetection:
Se = 58.0% (40.0–73.0%)
Sp = 86.0% (80.0–90.0%)
Recurrence:
Se = 25.0% (0.63–80.6%) (early GC),
45.5% (30.4–61.1%) (advanced GC)
Sp = 88.6% (84.6–92.0%) (early GC),
84.6% (76.9–90.4%) (advanced GC)
Low sensitivity in early stages, poor specificity and low positive predictive valuePhase 2[50,51,52]
SCC-antigen (Squamous Cell Carcinoma Ag): ELISA; Class: Protein (squamous marker)Esophageal squamous carcinomaResponse evaluation; recurrence surveillance in ESCCDetection of EC:
Se = 35.1% (32.0–38.3%)
Sp = 95.4 (93.8–96.7%);
Recurrence:
Se = 26.8% (14.2–42.9%),
Sp = Not reported;
elevated SCC-antigen associated with poor OS
Low sensitivity and lack of diagnostic specificity due to elevation in non-malignant conditionsPhase 2[44,53]
Pepsinogen I/II + H. pylori serology (“ABC” test) ELISA; Class: Protein enzymes (PGI, PGII) + antibodyGastric (screening for risk)Screening risk stratification (detects atrophic gastritis)GC (detection):
Se = 87.9% (71.8–96.6%),
Sp = 50.8% (37.9–63.6%)
Low sensitivity, poor performance for early cancer, less effective for cancers of cardia and pylorus, and affected by the use of PPIs and presence of certain H pylori strainsPhase 2[54]
Methylated Reprimo (RPRM) DNA (MSP assay) Class: ctDNA (methylated tumor DNA)Gastric adenocarcinomaEarly detection; post-op surveillanceGC (detection):
Se = 65.0% (53.5–75.3%),
Sp = 75.9% (73.2–78.5%)
Heterogeneity in methylation levels across different stages of cancer, potential for false positives, and difficulty in distinguishing early-stage lesions from normal tissuePhase 2[55]
Circulating Tumor DNA—personalized panel (NGS) Class: ctDNA (mutations, INDELs)Esophagus (EAC, ESCC); GastricMRD detection post-surgery; relapse surveillanceEsophagus/Gastric (Natera MRD detection):
Se = 85.7% (69.7–95.2%)
Sp = 95.5% (88.9–98.8%)
Insufficient sensitivity for very low ctDNA concentrations and high technical variabilityPhase 2[56]
Serum DSG2 (Desmoglein-2) ELISA; Class: Protein (adhesion molecule)ESCC; EGJ adenocarcinomaDiagnostic/prognostic marker (ESCC)ESCC (detection):
Se = 58.2% (43.2–70.8%)
Sp = 84.7% (73.0–92.8%)
EJA (detection):
Se = 29.2% (20.6–39.5%)
Sp = 90.2% (79.1–96.0%)
Low specificity, varied diagnostic accuracy based on cancer subtype and suboptimal sensitivity for early stagePhase 1[57]
Multi-TAA autoantibody panel Muliplex Immunoassay; Class: AutoantibodiesEsophageal adenocarcinoma (Barrett’s)Risk stratify Barrett’s progressionDetection:
Se = 53.5–64.0%
Sp = 87.0–93.7%
Inter-assay variability, lack of standardization across different multiplex platforms, and the inherent variability of autoantibody responsesPhase 2[58]
D-mannose (serum metabolite): LC-MS; Class: Metabolite (sugar)esophageal adenocarcinomaPrognostic biomarker (EAC)Low levels associated with poor prognosis; insufficient literature to report performance metricsLack of standardized clinical assays and high pre-analytical variabilityPhase 1[59]
Abbreviations: CEA: carcinoembryonic antigen, ELISA: enzyme-linked immunosorbent assay, EC: esophageal cancer; GC: gastric cancer, CA: carbohydrate antigen, SCC: squamous cell carcinoma, ESCC: esophageal squamous cell carcinoma, PG: pepsinogen. RPRM: Reprimo, DNA: deoxyribonucleic acid, NGS: next-generation sequencing, ctDNA: circulating tumor DNA, MRD: minimal residual disease, DSG2: desmoglein-2, EGJ: esophagogastric junction, EJA: esophagogastric junction adenocarcinoma, TAA: tumor-associated antigen.
Several completed clinical trials have significantly advanced our understanding and application of blood-based biomarkers in the surveillance and management of gastric and esophageal cancers, establishing their utility across various clinical scenarios(Table 2). One notable achievement comes from the prospective multicenter NCT05431621 (China) study, which successfully developed and validated “GutSeer,” a multi-analyte ctDNA panel incorporating methylation and fragmentomics for the non-invasive detection of multiple GI cancers, including gastric and esophageal, demonstrating high diagnostic performance in an independent cohort (NCT05431621). Beyond initial detection, the predictive power of these markers for metastasis and recurrence has been rigorously investigated. For instance, NCT02159339 (Korea) identified specific DNA methylation patterns (GFRA1m and ZNF382m) as potential biomarkers for predicting gastric cancer metastasis. At the same time, NCT04830618 (Korea) highlighted the predictive value of methylation of the c-mos proto-oncogene (MOS) for metachronous recurrence following endoscopic resection of gastric neoplasms (NCT02159339, NCT04830618).
Furthermore, ctDNA has emerged as a powerful tool for detecting MRD and recurrence monitoring. However, using ctDNA for early cancer detection and surveillance faces several challenges. Cancer cells release minimal amounts of DNA into the bloodstream, making it difficult to distinguish tumor-specific mutations from the abundance of normal DNA. A negative ctDNA result does not always confirm the absence of cancer; ctDNA levels may be below the limit of detection, or the specific mutation being tested for may not be present in the patient’s tumor. Non-cancerous conditions, such as clonal hematopoiesis of indeterminate potential (CHIP) the accumulation of age-related mutations in blood cells and other inflammatory conditions can introduce mutations that mimic cancer-related mutations, leading to false-positive results [60]. Finally, ctDNA has a short half-life, and its degradation during or after sample collection can further complicate detection efforts. Studies such as NCT02887612 (China), an impactful retrospective cohort study from China, demonstrated that postoperative ctDNA positivity in resected gastric cancer patients serves as a robust predictor for recurrence and inferior survival outcomes, often providing an earlier indication of relapse than conventional imaging NCT02887612, (tumor-informed ctDNA). Similar prognostic implications of post-treatment ctDNA dynamics have been observed in esophageal cancers, including ESCC and EAC, across various international cohorts (China, ESCC ctDNA), A United Kingdom cohort study [60] without a registered NCT identifier similarly reported that postoperative ctDNA positivity in esophageal adenocarcinoma predicted relapse within 12 months, as evidenced by NCT03425058 (China), which established that dynamic changes in ctDNA and circulating tumor cells (CTCs) during neoadjuvant chemotherapy for gastric adenocarcinoma correlate well with pathological and radiological response assessments (NCT03425058). This dynamic monitoring allows for personalized therapeutic adjustments, as demonstrated by trials such as NCT02674373 (France), which show that early clearance of ctDNA during or after neoadjuvant therapy correlates with improved patient outcomes in GE adenocarcinoma. Conversely, persistent positivity indicates poorer prognoses (NCT02674373). Finally, in the context of neoadjuvant therapy for ESCC, the NCT04005170 (China) study revealed that ctDNA negativity during or post-chemoradiotherapy combined with immunotherapy is associated with higher complete clinical response rates and better survival outcomes, highlighting the potential for biomarker-guided treatment de-escalation or intensification based on response dynamics (NCT04005170). Notably, a multi-institutional phase IB trial (NCT03044613) assessing the safety and feasibility of neoadjuvant nivolumab or nivolumab-relatlimab in resectable GE cancer patients investigated the association of serial ctDNA time points with clinical outcomes [61]. This study demonstrated that negative ctDNA preoperatively, postoperatively, and after immune checkpoint inhibition induction was associated with significantly longer recurrence-free survival and overall survival [61]. Collectively, these completed trials highlight the transformative potential of blood-based biomarkers to refine surveillance strategies, personalized treatment, and ultimately enhance the clinical outcomes for patients with GE cancers.
Table 2. Completed Clinical Trials Evaluating Blood-Based Biomarkers in Gastroesophageal Cancers.
Table 2. Completed Clinical Trials Evaluating Blood-Based Biomarkers in Gastroesophageal Cancers.
Trial (Study) & ID, CountryDesign
(Biomarkers)
Sample SizeStudy AimEndpointsStatus/Key FindingsReferences
NCT05431621; ChinaProspective multicenter case–control;
Biomarkers: “GutSeer” (1656-locus methylation + fragmentomics ctDNA panel)
Training/validation:
1057 cancer cases vs. 1415 controls;
Testing cohort: 846 patients
To develop GutSeer, a blood-based assay using DNA methylation and fragmentomics for multi-GI cancer detection (colorectal, esophagus, gastric, pancreas, and liver)Diagnostic performance of GutSeer assay for detecting GI cancers, measured by sensitivity, specificity, and AUC in an independent validation cohort.Completed; Overall GI cancer cohort: AUC = 92.1% Se = 81.5%, Sp = 94.4%;
Gastric: Se = 90.5%
Esophagus: Se = 65.2%
[62]
NCT03425058; ChinaProspective single center;
Biomarkers: dMMR/MSI status with dynamic evaluation of CTC and ctDNA
50To verify the value of ctDNA and CTC as biomarkers for tumor response in the neoadjuvant chemotherapy (nCRT) treatment of locally advance gastric adenocarcinoma.Concordance and accuracy of response evaluation results determined by ctDNA, CTCs compared with imaging and serum tumor biomarkers (CEA, CA19-9, CA72-4)Completed; ctDNA and CTC alteration during neoadjuvant therapy is consistent with conventional histopathological grading and radiological response assessment.[63]
NCT05227261, VietnamProspective multicenter;
Biomarkers: SPOT-MAS multimodal ctDNA panel (methylation, fragmentomics, copy number, end motif)
9057To validate the clinical utility of a multimodal non-invasive ctDNA-based MCED test, SPOT-MASPPV, NPV, sensitivity, and specificity of the blood ctDNA test in early detection of cancers (breast, lung, gastric, liver, colorectal)Completed; PPV = 39.5%, NPV = 99.9%, Se = 70.7%,
Sp = 99.7%; performance metrics for detecting various cancer types at 12 month follow up
[13]
NCT02159339, KoreaProspective cohort;
Biomarkers: GFRA1, SRF, ZNF382 methylation alterations. P16 and E-cadherin status as well.
198To evaluate the feasibility of predicting GC metastasis using CDH1, GFRA1, P16 and ZNF382 DNA methylation as biomarkers.HR, PPV and NPV of recurrence/metastasis of gastric cancer based on different methylation status Completed; GFRA1m and ZNF382m are potential biomarkers for the prediction of pN0M0 GC metastasis[64]
NCT04830618, KoreaProspective cohort;
Biomarkers: MOS methylation
294 overall; 123 gastric cancer vs. 171 gastric dysplasiaTo evaluate if MOS methylation can be used to predict metachronous recurrence after endoscopic resection of gastric neoplasms.MOS methylation for prediction of metachronous recurrence at least 1 year after diagnosisCompleted; MOS methylation predictive (adjusted HR = 4.76) for metachronous recurrence after endoscopic resection for gastric cancer.
Se = 80.0%, sp = 53.2%,
[65]
NCT02887612, ChinaProspective cohort; Biomarkers: ctDNA (targeted sequencing panel of 425 cancer-related genes)100Predictive value of ctDNA in Early and intermediate-stage gastric cancerPositive Predictive Value; The proportions of patients with positive serum ctDNA that have postoperative relapseCompleted; postoperative positive ctDNA, HR = 2.74 for recurrence vs. post-ACT positive ctDNA, HR = 15.0.
Post ACT ctDNA, se = 77.8%, sp = 90.6% for recurrence.
[66]
NCT02674373, FranceProspective cohort;
Biomarkers: ctDNA
82ctDNA to predict response and risk stratification in gastric or GE adenocarcinomaPFS, OS and tumor response rateCompleted; ctDNA + ve during NAT (HR = 6.2), post NAT (HR = 5.3), and after surgery (HR = 12.9) associated with worst outcomes. Early ctDNA clearance during NAT associated with better outcomes[67]
No NCT number, ChinaProspective cohort;
Biomarkers: tumor-informed ctDNA (NGS)
46To evaluate MRD detection by ctDNA and its association with clinical outcome in resected gastric cancerDFS and OSCompleted; ctDNA + ve in post-op period associated with DFS and OS (HR  =  14.78 and HR = 7.66, respectively) and preceded radiographic recurrence by a median of 6 months. [68]
NCT04005170, ChinaInterventional Phase 2 open label;
Biomarkers: tumor naïve ctDNA (NGS)
42To evaluate the efficacy and safety of the combination of toripalimab (an anti-PD-1 antibody) combined with definitive CRT in locally advanced ESCCcCR, OS, PFS, duration of response and QOLCompleted; ctDNA -ve patients had a high a cCR to those with detectable ctDNA during CRT83% vs. 39%) or post CRT (78% vs. 30%). ctDNA + ve, shorter PFS and OS.[69]
NCT04460066, NCT05543057, ChinaProspective cohort;
Biomarkers: ctDNA
89To develop a MRD profiling approach with enhanced sensitivity and specificity for detecting minimal tumor DNA from cfDNA in ESCCpCR Completed; MRD -ve associated with pCR in neoadjuvant, surgical, and adjuvant therapy cohort whereas MRD + ve was associated with non-pCR. All MRD -ve patients stayed progression free while 23/26 MRD + ve developed progression. Similar MRD results for radiotherapy cohort[70]
No NCT number, ChinaRetrospective cohort;
Biomarkers: ctDNA (NGS of 77 genes)
147Clinical utility of longitudinal ctDNA as a prognostic biomarker in ESCCOS and PFSCompleted; For curative surgical resection, high ctDNA (HR = 7.84) and (HR = 5.71) ctDNA alterations associated with poor OS. NAT group, post NAT ctDNA (HR = 3.16) alterations associated with poor PFS. [71]
No NCT number, United StatesRetrospective cohort;
Biomarkers: ctDNA (CAPP-Seq)
45To evaluate whether ctDNA analysis can predict recurrence in patients with localized ESCA earlier than standard-of-care imagingDistant metastases, OS and progressionCompleted; Detection of ctDNA was associated with tumor progression, metastasis, and disease-specific survival.[72]
No NCT number, United KingdomProspective cohort;
Biomarkers: ctDNA (77 gene panel)
97Prognostic potential of ctDNA dynamics in EAC, taking into account CHIPRecurrenceCompleted; ctDNA in plasma following surgery for EAC is prognostic for relapse[60]
No NCT number,
Netherlands, Sweden, and Denmark
Retrospective cohort;
Biomarkers: ctDNA
42To detect ctDNA alterations after preoperative chemotherapy and after surgery in patients with resectable gastric cancerRecurrenceCompleted; ctDNA is a predictive biomarker of patient outcome to perioperative cancer therapy and surgical resection in patients with gastric cancer.[73]
NCT03044613, United StatesPhase IB, open-label, multicenter trial;
Biomarkers: ctDNA
32To assess the safety and feasibility of nivolumab +/− relatlimab prior to chemoradiation with II/III gastro/esophageal Cancer Safety, feasibility, OS, RFS, MPR and pCR. ctDNA association with RFS and OS. Completed; undetectable ctDNA post-ICI induction, preoperatively and postoperatively had a significantly longer RFS and OS[61]
Abbreviation: NCT: National Clinical Trial identifier, ctDNA: circulating tumor DNA, GI: gastrointestinal, AUC: area under the curve, dMMR/MSI: deficient mismatch repair/microsatellite instability, CTC: circulating tumor cell, nCRT: neoadjuvant chemoradiotherapy, CEA: carcinoembryonic antigen, CA: carbohydrate antigen, SPOT-MAS: Screening for the Presence of Tumor by Methylation and Size, MCED: multi-cancer early detection, PPV: positive predictive value, NPV: negative predictive value, GFRA1: GDNF family receptor alpha-1, SRF: serum response factor, ZNF382: zinc finger protein 382, GC: gastric cancer, CDH1: cadherin-1, MOS: median overall survival, HR: hazard ratio, ACT: adjuvant chemotherapy, PFS: progression-free survival, OS: overall survival, NAT: neoadjuvant therapy, NGS: next-generation sequencing, PD-1: programmed cell death protein 1, DFS: disease-free survival, CRT: chemoradiotherapy, ESCC: esophageal squamous cell carcinoma, cCR: clinical complete response, QOL: quality of life, MRD: minimal residual disease, CAPP: Cancer Personalized Profiling, ESCA: esophageal carcinoma, CHIP: clonal hematopoiesis of indeterminate potential, EAC: esophageal adenocarcinoma, RFS: recurrence-free survival, MPR: major pathological response, pCR: pathological complete response, ICI: immune checkpoint inhibitor.

3. Ongoing Trials

Across ongoing trials, ctDNA remains the dominant analyte evaluated for minimal residual disease detection and treatment-response monitoring. Key endpoints include ctDNA-defined recurrence-free survival and correlation with imaging. Several trials explore methylation-based and multi-analyte panels integrating cfDNA, cfRNA, and extracellular-vesicle markers. Collectively, these studies aim to establish actionable thresholds for recurrence risk and assess whether serial liquid-biopsy monitoring can complement or reduce imaging-based surveillance. Many of these studies focus on ctDNA as a tool for detecting minimal residual disease (MRD), predicting recurrence, and monitoring treatment response. Other trials are exploring the potential of ctDNA in specific clinical scenarios, such as NCT06498752 (China) (Table 3), which is evaluating whether ctDNA MRD status after radical radiotherapy can guide consolidation therapy with PD-1 inhibitors in patients with esophageal squamous cell carcinoma (ESCC). These ongoing trials are expected to provide valuable insights into the role of blood-based biomarkers in improving the management of GE cancers. The ongoing trials highlight several potential applications of blood-based biomarkers in the surveillance of GE cancers. ctDNA is being extensively investigated for MRD detection post-treatment in trials such as NCT05029869 and NCT06893133, to identify high-risk patients for adjuvant therapy or closer monitoring, and for early treatment efficacy indication through ctDNA clearance. Biomarkers, including ctDNA, are also being explored to predict treatment response in trials such as NCT04053725 and NCT06662110, with the potential to inform personalized treatment decisions. Longitudinal monitoring of biomarkers such as ctDNA and CTCs, as seen in NCT02610218, could provide real-time insights into disease progression and treatment response, allowing for earlier recurrence detection than traditional imaging. Furthermore, trials such as NCT05366881 and NCT07035587 are developing blood-based tests for early cancer detection, potentially improving outcomes through earlier intervention.

4. Translational Significance and Future Directions

The integration of blood-based biomarkers promises to transform GE surveillance, enabling earlier recurrence detection and less invasive monitoring (Figure 2). This strategy utilizes multi-analyte biomarker panels in high-risk patients or with Barrett’s esophagus. A positive biomarker result triggers imaging and/or endoscopy, while AI integrates biomarker data with imaging and patient history for personalized risk assessment. Confirmed recurrence prompts treatment, whereas negative results guide continued routine surveillance, potentially with serial blood tests. This approach, by reducing unnecessary invasive procedures, aims to personalize follow-up and significantly improve patient outcomes. Recent advances integrate genomics, epigenomics, proteomics, and metabolomics into multi-omics liquid biopsy assays. Multi-cancer early detection (MCED) tests such as GutSeer and SPOT-MAS exemplify this approach, detecting several GI cancers in a single assay. Despite encouraging accuracy, real-world adoption will depend on cost-effectiveness, laboratory standardization, and reimbursement frameworks. Modeling studies suggest that cost per recurrence detected must fall below imaging-based surveillance to achieve widespread implementation. Future directions include seamless integration with AI-enhanced imaging, MRD-guided therapy escalation/de-escalation, risk-stratified screening, and composite biomarker panels. Despite challenges in standardization and sensitivity, blood-based surveillance offers a more convenient and compliant approach, heralding a future where a simple blood draw guides timely curative action.

5. Conclusions

Liquid biopsy shows strong potential for clinical integration, but its widespread adoption will depend on large, prospective validation and demonstration of cost-effectiveness in real-world settings. Blood-based surveillance biomarkers have the potential to catch recurrences earlier (when they are still curable), reduce unnecessary invasive procedures, and personalize follow-up strategies. Combined with powerful imaging modalities and AI analytics, they promise a future where no patient with GE cancers falls through the cracks of surveillance. Artificial intelligence-driven classifiers increasingly integrate cfDNA fragmentomics, methylation, and proteomic signals using large public datasets (TCGA, ICGC) and proprietary multi-cancer training cohorts. However, validation across independent, prospectively collected cohorts remains limited, and regulatory frameworks for AI-based clinical decision tools are still evolving. Achieving this vision will require ongoing interdisciplinary collaboration and validation in clinical trials, but the foundation has been laid by the advances summarized in this review. The fight against GE cancers is poised to leverage these minimally invasive tools to improve patient outcomes and quality of life, fulfilling the urgent need that clinicians and patients alike have long recognized.

Author Contributions

Conceptualization, N.D. and A.H.Z.; Methodology, C.S. imaging and writing first draft, M.A., E.E.G. and A.G.; Data Curation, H.Y.P., A.F.K. and A.O.; Writing—Original Draft, N.D.; Writing—Review & Editing, D.L.B., P.L.W. and A.H.Z.; Supervision, A.H.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Qi, L.; Sun, M.; Liu, W.; Zhang, X.; Yu, Y.; Tian, Z.; Ni, Z.; Zheng, R.; Li, Y. Global esophageal cancer epidemiology in 2022 and predictions for 2050: A comprehensive analysis and projections based on GLOBOCAN data. Chin. Med. J. 2024, 137, 3108–3116. [Google Scholar] [CrossRef]
  3. Alcasid, N.J.; Fink, D.; Banks, K.C.; Susai, C.J.; Barnes, K.; Wile, R.; Sun, A.; Patel, A.; Ashiku, S.; Velotta, J.B. The Impact of Diagnostic Laparoscopy on Upstaging Patients with Siewert II and III Gastroesophageal Junction (GEJ) Cancer. Ann. Surg. Oncol. 2024, 32, 258–264. [Google Scholar] [CrossRef]
  4. Nassour, I.; Fullington, H.; Hynan, L.S.; Yopp, A.C.; Augustine, M.M.; Polanco, P.M.; Choti, M.A.; Mansour, J.C.; Wang, S.C.; Porembka, M.R. The Yield of Staging Laparoscopy in Gastric Cancer is Affected by Racial and Ethnic Differences in Disease Presentation. Ann. Surg. Oncol. 2017, 24, 1787–1794. [Google Scholar] [CrossRef] [PubMed]
  5. Schena, C.A.; Laterza, V.; De Sio, D.; Quero, G.; Fiorillo, C.; Gunawardena, G.; Strippoli, A.; Tondolo, V.; De’angelis, N.; Alfieri, S.; et al. The Role of Staging Laparoscopy for Gastric Cancer Patients: Current Evidence and Future Perspectives. Cancers 2023, 15, 3425. [Google Scholar] [CrossRef] [PubMed]
  6. Bandidwattanawong, C. Multi-disciplinary management of esophageal carcinoma: Current practices and future directions. Crit. Rev. Oncol. 2024, 197, 104315. [Google Scholar] [CrossRef]
  7. Scheck, M.K.; Hofheinz, R.D.; Lorenzen, S. HER2-Positive Gastric Cancer and Antibody Treatment: State of the Art and Future Developments. Cancers 2024, 16, 1336. [Google Scholar] [CrossRef] [PubMed]
  8. Jiao, X.; Wang, Y.; Wang, F.; Wang, X. Recurrence pattern and its predictors for advanced gastric cancer after total gastrectomy. Medicine 2020, 99, e23795. [Google Scholar] [CrossRef]
  9. Wen, X.; Pu, H.; Liu, Q.; Guo, Z.; Luo, D. Circulating Tumor DNA—A Novel Biomarker of Tumor Progression and Its Favorable Detection Techniques. Cancers 2022, 14, 6025. [Google Scholar] [CrossRef]
  10. Zhu, L.; Xu, R.; Yang, L.; Shi, W.; Zhang, Y.; Liu, J.; Li, X.; Zhou, J.; Bing, P. Minimal residual disease (MRD) detection in solid tumors using circulating tumor DNA: A systematic review. Front. Genet. 2023, 14, 1172108. [Google Scholar] [CrossRef]
  11. Labiano, I.; Huerta, A.E.; Arrazubi, V.; Hernandez-Garcia, I.; Mata, E.; Gomez, D.; Arasanz, H.; Vera, R.; Alsina, M. State of the Art: ctDNA in Upper Gastrointestinal Malignancies. Cancers 2023, 15, 1379. [Google Scholar] [CrossRef]
  12. Wang, J.; Huang, J.; Hu, Y.; Guo, Q.; Zhang, S.; Tian, J.; Niu, Y.; Ji, L.; Xu, Y.; Tang, P.; et al. Terminal modifications independent cell-free RNA sequencing enables sensitive early cancer detection and classification. Nat. Commun. 2024, 15, 156. [Google Scholar] [CrossRef]
  13. Nguyen, L.H.D.; Nguyen, T.H.H.; Van Le, H.; Bui, V.Q.; Nguyen, L.H.; Pham, N.H.; Phan, T.H.; Nguyen, H.T.; Tran, V.S.; Bui, C.V. Prospective validation study: A non-invasive circulating tumor DNA-based assay for simultaneous early detection of multiple cancers in asymptomatic adults. BMC Med. 2025, 23, 90. [Google Scholar] [CrossRef]
  14. Roy, S.; Kanda, M.; Nomura, S.; Zhu, Z.; Toiyama, Y.; Taketomi, A.; Goldenring, J.; Baba, H.; Kodera, Y.; Goel, A. Diagnostic efficacy of circular RNAs as noninvasive, liquid biopsy biomarkers for early detection of gastric cancer. Mol. Cancer 2022, 21, 42. [Google Scholar] [CrossRef] [PubMed]
  15. Ortiz-Quintero, B. Extracellular MicroRNAs as Intercellular Mediators and Noninvasive Biomarkers of Cancer. Cancers 2020, 12, 3455. [Google Scholar] [CrossRef] [PubMed]
  16. Roșu, M.C.; Ardelean, A.; Moldovan, S.D.; Faur, F.I.; Nesiu, A.; Totoloci, B.D. The importance of CA 72-4 and CA 19-9 dosing in gastric cancer. J. Med. Life 2023, 16, 186–188. [Google Scholar] [CrossRef]
  17. Kotzev, A.I.; Draganov, P.V. Carbohydrate Antigen 19-9, Carcinoembryonic Antigen, and Carbohydrate Antigen 72-4 in Gastric Cancer: Is the Old Band Still Playing? Gastrointest. Tumors 2018, 5, 1–13. [Google Scholar] [CrossRef]
  18. Căinap, C.; Nagy, V.; Gherman, A.; Cetean, S.; Laszlo, I.; Constantin, A.-M.; Căinap, S. Classic Tumor Markers in Gastric Cancer. Current Standards and Limitations. Med. Pharm. Rep. 2015, 88, 111–115. [Google Scholar] [CrossRef] [PubMed]
  19. Ma, Y.; Qiu, C.; Wang, B.; Zhang, X.; Wang, X.; Aguilera, R.J.; Zhang, J.-Y. Autoantibody against Tumor-Associated Antigens as Diagnostic Biomarkers in Hispanic Patients with Hepatocellular Carcinoma. Cells 2022, 11, 3227. [Google Scholar] [CrossRef]
  20. de Jonge, H.; Iamele, L.; Maggi, M.; Pessino, G.; Scotti, C. Anti-Cancer auto-antibodies: Roles, applications and open issues. Cancers 2021, 13, 813. [Google Scholar] [CrossRef]
  21. Bizzaro, N.; Antico, A.; Villalta, D. Autoimmunity and gastric cancer. Int. J. Mol. Sci. 2018, 19, 377. [Google Scholar] [CrossRef]
  22. Desmetz, C.; Mange, A.; Maudelonde, T.; Solassol, J. Autoantibody signatures: Progress and perspectives for early cancer detection. J. Cell. Mol. Med. 2011, 15, 2013–2024. [Google Scholar] [CrossRef]
  23. Wang, Y.; Jiao, Y.; Ding, C.-M.; Sun, W.-Z. The role of autoantibody detection in the diagnosis and staging of lung cancer. Ann. Transl. Med. 2021, 9, 1673. [Google Scholar] [CrossRef] [PubMed]
  24. Ju, S.; Chen, C.; Zhang, J.; Xu, L.; Zhang, X.; Li, Z.; Chen, Y.; Zhou, J.; Ji, F.; Wang, L. Detection of circulating tumor cells: Opportunities and challenges. Biomark. Res. 2022, 10, 58. [Google Scholar] [CrossRef] [PubMed]
  25. Shi, Y.; Ge, X.; Ju, M.; Zhang, Y.; Di, X.; Liang, L. Circulating Tumor Cells in Esophageal Squamous Cell Carcinoma—Mini Review. Cancer Manag. Res. 2021, 13, 8355–8365. [Google Scholar] [CrossRef] [PubMed]
  26. Yang, C.; Zhang, N.; Wang, S.; Shi, D.; Zhang, C.; Liu, K.; Xiong, B. Wedge-Shaped microfluidic chip for circulating tumor cells isolation and its clinical significance in gastric cancer. J. Transl. Med. 2018, 16, 139. [Google Scholar] [CrossRef]
  27. Kanayama, M.; Yoneda, K.; Kuwata, T.; Mori, M.; Manabe, T.; Oyama, R.; Matsumiya, H.; Takenaka, M.; Kuroda, K. Enhanced capture system for mesenchymal-type circulating tumor cells using a polymeric microfluidic device ‘CTC-Chip’incorporating cell-surface vimentin. Oncol. Rep. 2024, 52, 156. [Google Scholar] [CrossRef]
  28. Doyle, L.; Wang, M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef]
  29. Wang, Z.; Wang, Q.; Qin, F.; Chen, J. Exosomes: A promising avenue for cancer diagnosis beyond treatment. Front. Cell Dev. Biol. 2024, 12, 1344705. [Google Scholar] [CrossRef]
  30. Tang, S.; Cheng, J.; Yao, Y.; Lou, C.; Wang, L.; Huang, X.; Zhang, Y. Combination of Four Serum Exosomal MiRNAs as Novel Diagnostic Biomarkers for Early-Stage Gastric Cancer. Front. Genet. 2020, 11, 237. [Google Scholar] [CrossRef]
  31. Li, L.; Sun, Y. Circulating tumor DNA methylation detection as biomarker and its application in tumor liquid biopsy: Advances and challenges. Medcomm 2024, 5, e766. [Google Scholar] [CrossRef]
  32. Geissler, F.; Nesic, K.; Kondrashova, O.; Dobrovic, A.; Swisher, E.M.; Scott, C.L.; Wakefield, M.J. The role of aberrant DNA methylation in cancer initiation and clinical impacts. Ther. Adv. Med. Oncol. 2024, 16, 17588359231220511. [Google Scholar] [CrossRef]
  33. Saliminejad, K.; Soleymani Fard, S.; Khorram Khorshid, H.R.; Yaghmaie, M.; Mahmoodzadeh, H.; Mousavi, S.A.; Ghaffari, S.H. Methylation Analysis of P16, RASSF1A, RPRM, and RUNX3 in Circulating Cell-Free DNA for Detection of Gastric Cancer: A Validation Study. Avicenna J. Med. Biotechnol. 2020, 12, 99–106. [Google Scholar] [PubMed]
  34. Basharat, S.; Horton, J. Emerging Multi-Cancer Early Detection Technologies. Can. J. Health Technol. 2022, 2. [Google Scholar] [CrossRef]
  35. Loomans-Kropp, H.A.; Song, Y.; Gala, M.; Parikh, A.R.; Van Seventer, E.E.; Alvarez, R.; Hitchins, M.P.; Shoemaker, R.H.; Umar, A. Methylated Septin9 (mSEPT9): A Promising Blood-Based Biomarker for the Detection and Screening of Early-Onset Colorectal Cancer. Cancer Res. Commun. 2022, 2, 90–98. [Google Scholar] [CrossRef]
  36. Zhu, L.; Li, X.; Yuan, Y.; Dong, C.; Yang, M. APC Promoter Methylation in Gastrointestinal Cancer. Front. Oncol. 2021, 11, 653222. [Google Scholar] [CrossRef]
  37. Xu, Y.; Wang, Z.; Pei, B.; Wang, J.; Xue, Y.; Zhao, G. DNA methylation markers in esophageal cancer. Front. Genet. 2024, 15, 1354195. [Google Scholar] [CrossRef]
  38. Nannini, G.; Meoni, G.; Amedei, A.; Tenori, L. Metabolomics profile in gastrointestinal cancers: Update and future perspectives. World J. Gastroenterol. 2020, 26, 2514–2532. [Google Scholar] [CrossRef] [PubMed]
  39. Du, L.; Li, S.; Xiao, X.; Li, J.; Sun, Y.; Ji, S.; Jin, H.; Hua, Z.; Ma, J.; Wang, X.; et al. Metabolomic profiling of plasma reveals potential biomarkers for screening and early diagnosis of gastric cancer and precancerous stages. MedComm–Oncol. 2023, 2, e32. [Google Scholar] [CrossRef]
  40. Benfatto, S.; Sill, M.; Jones, D.T.W.; Pfister, S.M.; Sahm, F.; von Deimling, A.; Capper, D.; Hovestadt, V. Explainable artificial intelligence of DNA methylation-based brain tumor diagnostics. Nat. Commun. 2025, 16, 1787. [Google Scholar] [CrossRef]
  41. Sahoo, K.; Lingasamy, P.; Khatun, M.; Sudhakaran, S.L.; Salumets, A.; Sundararajan, V.; Modhukur, V. Artificial Intelligence in cancer epigenomics: A review on advances in pan-cancer detection and precision medicine. Epigenetics Chromatin 2025, 18, 35. [Google Scholar] [CrossRef] [PubMed]
  42. Chung, Y.; Kim, J.S.; Youk, H.-J.; Kim, H.; Hwang, S.-H.; Oh, H.-B.; Ko, D.-H. Relative immunogenicity of blood group antigens: First report in a Korean population. Transfus. Apher. Sci. 2022, 62, 103585. [Google Scholar] [CrossRef] [PubMed]
  43. Xing, H.; Wang, J.; Wang, Y.; Tong, M.; Hu, H.; Huang, C.; Li, D. Diagnostic Value of CA 19-9 and Carcinoembryonic Antigen for Pancreatic Cancer: A Meta-Analysis. Gastroenterol. Res. Pract. 2018, 2018, 8704751. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, J.; Zhu, Z.; Liu, Y.; Jin, X.; Xu, Z.; Yu, Q.; Li, K. Diagnostic Value of Multiple Tumor Markers for Patients with Esophageal Carcinoma. PLoS ONE 2015, 10, e0116951. [Google Scholar] [CrossRef]
  45. Bagaria, B.; Sood, S.; Sharma, R.; Lalwani, S. Comparative study of CEA and CA19-9 in esophageal, gastric and colon cancers individually and in combination (ROC curve analysis). Cancer Biol. Med. 2013, 10, 148–157. [Google Scholar] [CrossRef]
  46. Clark, G.W.; Ireland, A.P.; Hagen, J.A.; Collard, J.-M.; Peters, J.H.; DeMeester, T.R. Carcinoembryonic antigen measurements in the management of esophageal cancer: An indicator of subclinical recurrence. Am. J. Surg. 1995, 170, 597–601. [Google Scholar] [CrossRef]
  47. Zhang, R.; Chen, X.; Chen, G.; Zhao, Z.; Zhang, F.; Chen, Z.; Wei, C.; Lin, J.; Liu, Z.; Liang, Y.; et al. Novel surveillance protocol for gastric cancer based on CEA: A high-volume multi-center study. BMC Cancer 2025, 25, 1396. [Google Scholar] [CrossRef]
  48. Liang, Y.; Wang, W.; Fang, C.; Raj, S.S.; Hu, W.-M.; Li, Q.-W.; Zhou, Z.-W. Clinical significance and diagnostic value of serum CEA, CA19-9 and CA72-4 in patients with gastric cancer. Oncotarget 2016, 7, 49565–49573. [Google Scholar] [CrossRef]
  49. Shibata, C.; Nakano, T.; Yasumoto, A.; Mitamura, A.; Sawada, K.; Ogawa, H.; Miura, T.; Ise, I.; Takami, K.; Yamamoto, K.; et al. Comparison of CEA and CA19-9 as a predictive factor for recurrence after curative gastrectomy in gastric cancer. BMC Surg. 2022, 22, 213. [Google Scholar] [CrossRef]
  50. Wang, H.; Jin, W.; Wan, C.; Zhu, C. Diagnostic value of combined detection of CA72-4, CA19-9, and carcinoembryonic antigen comparing to CA72-4 alone in gastric cancer: A systematic review and meta-analysis. Transl. Cancer Res. 2022, 11, 848–856. [Google Scholar] [CrossRef]
  51. Hu, P.-J.; Chen, M.-Y.; Wu, M.-S.; Lin, Y.-C.; Shih, P.-H.; Lai, C.-H.; Lin, H.-J. Clinical Evaluation of CA72-4 for Screening Gastric Cancer in a Healthy Population: A Multicenter Retrospective Study. Cancers 2019, 11, 733. [Google Scholar] [CrossRef] [PubMed]
  52. Kim, D.H.; Oh, S.J.; Oh, C.A.; Choi, M.G.; Noh, J.H.; Sohn, T.S.; Bae, J.M.; Kim, S. The relationships between perioperative CEA, CA 19-9, and CA 72-4 and recurrence in gastric cancer patients after curative radical gastrectomy. J. Surg. Oncol. 2011, 104, 585–591. [Google Scholar] [CrossRef]
  53. Kawaguchi, H.; Ohno, S.; Miyazaki, M.; Hashimoto, K.; Egashira, A.; Saeki, H.; Watanabe, M.; Sugimachi, K. CYFRA 21-1 determination in patients with esophageal squamous cell carcinoma: Clinical utility for detection of recurrences. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2000, 89, 1413–1417. [Google Scholar] [CrossRef]
  54. In, H.; Adams, A.; Ho, S.; Sarkar, S.; Wang, F.; Oh, A.; Yun, D.J.; Gandhi, A.; Li, S.; Liu, H.; et al. Pepsinogen and Helicobacter pylori: Serum biomarkers for gastric cancer risk in a diverse United States population. Surg. Oncol. Insight 2024, 1, 100086. [Google Scholar] [CrossRef]
  55. Maturana, M.J.; Padilla, O.; Santoro, P.M.; Alarcón, M.A.; Olivares, W.; Blanco, A.; Armisen, R.; Garrido, M.; Aravena, E.; Barrientos, C.; et al. Methylated Reprimo Cell-Free DNA as a Non-Invasive Biomarker for Gastric Cancer. Int. J. Mol. Sci. 2025, 26, 3333. [Google Scholar] [CrossRef]
  56. Huffman, B.M.; Aushev, V.N.; Budde, G.L.; Chao, J.; Dayyani, F.; Hanna, D.; Botta, G.P.; Catenacci, D.V.; Maron, S.B.; Krinshpun, S.; et al. Analysis of Circulating Tumor DNA to Predict Risk of Recurrence in Patients With Esophageal and Gastric Cancers. JCO Precis. Oncol. 2022, 6, e2200420. [Google Scholar] [CrossRef]
  57. Liu, Y.-Q.; Chu, L.-Y.; Yang, T.; Zhang, B.; Zheng, Z.-T.; Xie, J.-J.; Xu, Y.-W.; Fang, W.-K. Serum DSG2 as a potential biomarker for diagnosis of esophageal squamous cell carcinoma and esophagogastric junction adenocarcinoma. Biosci. Rep. 2022, 42, BSR20212612. [Google Scholar] [CrossRef]
  58. Xu, Y.-W.; Peng, Y.-H.; Xu, L.-Y.; Xie, J.-J.; Li, E.-M. Autoantibodies: Potential clinical applications in early detection of esophageal squamous cell carcinoma and esophagogastric junction adenocarcinoma. World J. Gastroenterol. 2019, 25, 5049–5068. [Google Scholar] [CrossRef] [PubMed]
  59. Gu, J.; Liang, D.; Pierzynski, J.A.; Zheng, L.; Ye, Y.; Zhang, J.; Ajani, J.A.; Wu, X. D-Mannose: A Novel Prognostic Biomarker for Patients with Esophageal Adenocarcinoma. Carcinogenesis 2017, 38, 162–167. [Google Scholar] [CrossRef]
  60. Ococks, E.; Frankell, A.; Soler, N.M.; Grehan, N.; Northrop, A.; Coles, H.; Redmond, A.; Devonshire, G.; Weaver, J.; Hughes, C.; et al. Longitudinal tracking of 97 esophageal adenocarcinomas using liquid biopsy sampling. Ann. Oncol. 2021, 32, 522–532. [Google Scholar] [CrossRef] [PubMed]
  61. Kelly, R.J.; Landon, B.V.; Zaidi, A.H.; Singh, D.; Canzoniero, J.V.; Balan, A.; Hales, R.K.; Voong, K.R.; Battafarano, R.J.; Jobe, B.A.; et al. Neoadjuvant nivolumab or nivolumab plus LAG-3 inhibitor relatlimab in resectable esophageal/gastroesophageal junction cancer: A phase Ib trial and ctDNA analyses. Nat. Med. 2024, 30, 1023–1034. [Google Scholar] [CrossRef]
  62. Huang, A.; Guo, D.-Z.; Su, Z.-X.; Zhong, Y.-S.; Liu, L.; Xiong, Z.-G.; He, D.-L.; Yan, B.; Li, Q.-L.; Feng, Z.; et al. GUIDE: A prospective cohort study for blood-based early detection of gastrointestinal cancers using targeted DNA methylation and fragmentomics sequencing. Mol. Cancer 2025, 24, 163. [Google Scholar] [CrossRef]
  63. Fu, T. Monitoring patient-specific mutation in ctDNA and CTC for tumour response evaluation after neoadjuvant chemotherapy in locally advanced gastric cancer. Ann. Oncol. 2019, 30, v313–v314. [Google Scholar] [CrossRef]
  64. Liu, Z.; Cheng, X.; Zhang, L.; Zhou, J.; Deng, D.; Ji, J. A panel of DNA methylated markers predicts metastasis of pN0M0 gastric carcinoma: A prospective cohort study. Br. J. Cancer 2019, 121, 529–536. [Google Scholar] [CrossRef] [PubMed]
  65. Shin, C.M.; Kim, N.; Yoon, H.; Choi, Y.J.; Park, J.H.; Park, Y.S.; Lee, D.H. Aberrant DNA Methylation Maker for Predicting Metachronous Recurrence After Endoscopic Resection of Gastric Neoplasms. Cancer Res. Treat. 2022, 54, 1157–1166. [Google Scholar] [CrossRef]
  66. Yuan, S.; Nie, R.; Huang, Y.; Chen, Y.; Wang, S.; Sun, X.; Li, Y.; Liu, Z.; Chen, Y.; Yao, Y.; et al. Residual circulating tumor DNA after adjuvant chemotherapy effectively predicts recurrence of stage II-III gastric cancer. Cancer Commun. 2023, 43, 1312–1325. [Google Scholar] [CrossRef]
  67. Zaanan, A.; Didelot, A.; Broudin, C.; Laliotis, G.; Spickard, E.; Dutta, P.; Saltel-Fulero, A.; Sullo, F.G.; Pizzamiglio, M.; Mariani, A.; et al. Longitudinal circulating tumor DNA analysis during treatment of locally advanced resectable gastric or gastroesophageal junction adenocarcinoma: The PLAGAST prospective biomarker study. Nat. Commun. 2025, 16, 6815. [Google Scholar] [CrossRef] [PubMed]
  68. Yang, J.; Gong, Y.; Lam, V.K.; Shi, Y.; Guan, Y.; Zhang, Y.; Ji, L.; Chen, Y.; Zhao, Y.; Qian, F.; et al. Deep sequencing of circulating tumor DNA detects molecular residual disease and predicts recurrence in gastric cancer. Cell Death Dis. 2020, 11, 346. [Google Scholar] [CrossRef] [PubMed]
  69. Chen, B.; Liu, S.; Zhu, Y.; Wang, R.; Cheng, X.; Chen, B.; Dragomir, M.P.; Zhang, Y.; Hu, Y.; Liu, M. Predictive role of ctDNA in esophageal squamous cell carcinoma receiving definitive chemoradiotherapy combined with toripalimab. Nat. Commun. 2024, 15, 1919. [Google Scholar] [CrossRef]
  70. Yue, P.; Bie, F.; Zhu, J.; Gao, L.-R.; Zhou, Z.; Bai, G.; Wang, X.; Zhao, Z.; Xiao, Z.-F.; Li, Y.; et al. Minimal residual disease profiling predicts pathological complete response in esophageal squamous cell carcinoma. Mol. Cancer 2024, 23, 96. [Google Scholar] [CrossRef]
  71. Ng, H.Y.; Ko, J.M.Y.; Lam, K.O.; Kwong, D.L.W.; Lo, A.W.I.; Wong, I.Y.H.; Wong, C.L.Y.; Chan, S.Y.; Chan, K.K.; Law, T.T.; et al. Circulating Tumor DNA Dynamics as Prognostic Markers in Locally Advanced and Metastatic Esophageal Squamous Cell Carcinoma. JAMA Surg. 2023, 158, 1141–1150. [Google Scholar] [CrossRef] [PubMed]
  72. Azad, T.D.; Chaudhuri, A.A.; Fang, P.; Qiao, Y.; Esfahani, M.S.; Chabon, J.J.; Hamilton, E.G.; Yang, Y.D.; Lovejoy, A.; Newman, A.M.; et al. Circulating Tumor DNA Analysis for Detection of Minimal Residual Disease After Chemoradiotherapy for Localized Esophageal Cancer. Gastroenterology 2020, 158, 494–505.e6. [Google Scholar] [CrossRef] [PubMed]
  73. Leal, A.; van Grieken, N.C.T.; Palsgrove, D.N.; Phallen, J.; Medina, J.E.; Hruban, C.; Broeckaert, M.A.M.; Anagnostou, V.; Adleff, V.; Bruhm, D.C.; et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat. Commun. 2020, 11, 525. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The image outlines the different types of liquid biopsy assays used for monitoring Esophageal, Gastric Cancers, along with the specific biomarkers they detect and their clinical applications (Created in BioRender. Wagner, P. (2025) https://BioRender.com/4t32mvk, accessed on 25 October 2025) Abbreviations: ctDNA: circulating tumor DNA; EAC: esophageal adenocarcinoma; MRD: minimal residual disease; miRNA: microRNA; ESCC: esophageal squamous cell carcinoma; SCC: squamous cell carcinoma; CEA: carcinoembryonic antigen; CA: carbohydrate antigen; PGI: Pepsinogen I; PGII: Pepsinogen II.
Figure 1. The image outlines the different types of liquid biopsy assays used for monitoring Esophageal, Gastric Cancers, along with the specific biomarkers they detect and their clinical applications (Created in BioRender. Wagner, P. (2025) https://BioRender.com/4t32mvk, accessed on 25 October 2025) Abbreviations: ctDNA: circulating tumor DNA; EAC: esophageal adenocarcinoma; MRD: minimal residual disease; miRNA: microRNA; ESCC: esophageal squamous cell carcinoma; SCC: squamous cell carcinoma; CEA: carcinoembryonic antigen; CA: carbohydrate antigen; PGI: Pepsinogen I; PGII: Pepsinogen II.
Cancers 17 03552 g001
Figure 2. Proposed Algorithm for Integrating Blood-Based Biomarkers into GE Cancer Surveillance. This flowchart outlines a strategy for managing high-risk GE cancer patients, such as that post-resection or with Barrett’s esophagus. It integrates blood-based biomarker testing (preferably multi-analyte panels) with AI-powered personalized risk assessment, prompting further investigation (imaging and/or endoscopy with biopsy) for biomarker-positive cases to detect recurrence or disease. Negative biomarker results guide continued routine surveillance, potentially with repeat blood tests at defined intervals (Created in BioRender. Wagner, P. (2025) https://BioRender.com/gzun2g6, accessed on 25 October 2025). Abbreviations: AI: artificial intelligence, CT: computed tomography, PET/CT: positron emission tomography/computed tomography, SOC: standard of care, GE: gastroesophageal, MRD: minimal residual disease, cfDNA: cell-free DNA, ctDNA: circulating tumor DNA, EV: extracellular vesicle, miRNA: microRNA, NGS: next-generation sequencing, CA: carbohydrate antigen, CEA: carcinoembryonic antigen, ESCC: esophageal squamous cell carcinoma, EAC: esophageal adenocarcinoma.
Figure 2. Proposed Algorithm for Integrating Blood-Based Biomarkers into GE Cancer Surveillance. This flowchart outlines a strategy for managing high-risk GE cancer patients, such as that post-resection or with Barrett’s esophagus. It integrates blood-based biomarker testing (preferably multi-analyte panels) with AI-powered personalized risk assessment, prompting further investigation (imaging and/or endoscopy with biopsy) for biomarker-positive cases to detect recurrence or disease. Negative biomarker results guide continued routine surveillance, potentially with repeat blood tests at defined intervals (Created in BioRender. Wagner, P. (2025) https://BioRender.com/gzun2g6, accessed on 25 October 2025). Abbreviations: AI: artificial intelligence, CT: computed tomography, PET/CT: positron emission tomography/computed tomography, SOC: standard of care, GE: gastroesophageal, MRD: minimal residual disease, cfDNA: cell-free DNA, ctDNA: circulating tumor DNA, EV: extracellular vesicle, miRNA: microRNA, NGS: next-generation sequencing, CA: carbohydrate antigen, CEA: carcinoembryonic antigen, ESCC: esophageal squamous cell carcinoma, EAC: esophageal adenocarcinoma.
Cancers 17 03552 g002
Table 3. Liquid Biopsy-Based Clinical Trials for Surveillance in Gastroesophageal Cancers.
Table 3. Liquid Biopsy-Based Clinical Trials for Surveillance in Gastroesophageal Cancers.
Trial (Study) & ID, CountryDesign
(Biomarkers)
Sample SizeStudy AimEndpointsStatus/Key
Findings
NCT05029869; VietnamProspective observational;
Biomarkers: ctDNA
100Use of NGS to detect ctDNA in gastric cancer after surgerySensitivity/specificity of MRD detection using ctDNAOngoing (Active, not recruiting)
NCT06232395, ChinaProspective multicenter cohort:
Biomarkers: ctDNA
1197To develop and validate a new multi-target marker early detection and postoperative monitoring of gastric cancer Performance of the multi-target panel in diagnosis, detecting postoperative recurrence and metastasisOngoing, recruiting
NCT04993378, ChinaProspective cohort:
Biomarkers: extracellular vesicle (EV) protein signature “EV-score” derived from four EV proteins
40Validate whether the EV-score can predict and monitor immunotherapy outcomes in advanced gastric cancer, both at baseline and during treatmentPerformance of EV-score at baseline and longitudinally for prediction and monitoring of immunotherapeutic response (accuracy, sensitivity, specificity)Unknown
NCT04053725, ChinaProspective cohort;
Biomarkers: ctDNA
200Clinical utility of ctDNA in predicting the efficacy of immunotherapy for advanced gastric cancer.The proportions of patients with positive serum ctDNA that have postoperative recurrenceUnknown
NCT06662110,
China
Prospective cohort;
Biomarkers: PSRscore calculated based on baseline serum immune proteomics
206To validate the predictive value of systemic immune markers in predicting neoadjuvant treatment responses in advanced gastric cancer.Sensitivity and specificity of PSRscore in predicting tumor regression/objective response/PFS/OS after NAT Ongoing, recruiting
NCT03957564, ChinaInterventional phase II open label single group trial;
Biomarkers: CTC, ctDNA and cfDNA
40Clinical value of dynamic changes in CTC, ctDNA and cfDNA in NAT chemotherapy or and operation of resectable or locally advanced gastric or GEJ cancerComparison of biomarker dynamics with CT/RECIST responses and prognosis (e.g., recurrence, survival)Unknown
NCT06893133; ChinaProspective observational multicenter;
Biomarkers: personalized ctDNA-MRD
110Correlation between ctDNA-MRD status and tumor recurrence and metastasis in gastric cancer patients who have received neoadjuvant therapy followed by curative resectionSensitivity, Specificity, and Positive predictive value of ctDNA-MRD in predicting postoperative recurrenceOngoing (Active, not recruiting)
NCT01715233, United StatesPhase 2, single arm treatment trial;
Biomarkers: CHFR methylation
27To estimate and compare the response rates in metastatic GE patients treated with mDCF based on methylation status of CHFR.objective response rate (PR/SD/PD) stratified by CHFR methylationCompleted; results not reported
NCT06979895, ChinaProspective cohort;
Biomarkers: ctDNA multigene methylation panel
150Correlate methylation dynamics with treatment response for gastric cancerAssociation between changes in methylation and objective responseOngoing; not yet recruiting
NCT06335576, ChinaProspective single center cohort;
Biomarkers: serum proteomics panel
89Establish circulating proteomic subtypes of gastric cancer and explore their clinical applicabilityIdentification and reproducibility of serum-based proteomic subtypes (e.g., classification accuracy, subtype detection)Ongoing; not yet recruiting
NCT02610218, ChinaProspective cohort;
Biomarkers: ctDNA (HER2 amplification/mutations) + CTCs
124Evaluate whether changes in cfDNA levels and CTC counts correspond with therapeutic response to HER2-targeted therapy in metastatic HER2-positive gastric cancerConcordance of ctDNA/CTCs with radiologic treatment response, emergence of HER2 resistance, and detection of progressionUnknown
NCT04511559, ChinaProspective cohort;
Biomarkers: ctDNA methylation
540To describe ctDNA methylation profile in gastric cancer and demonstrate correlation between ctDNA methylation status and diagnosis and prognosisAnalysis of ctDNA methylation status and its correlation to early diagnosis and prognostic evaluation of gastric cancerUnknown
NCT05513144, ChinaProspective cohort;
Biomarkers: ctDNA
30To evaluate the use of next generation sequencing (NGS) to detect circulating tumor DNA in advanced HER2 negative gastric cancer patientsPrognostic molecular markers; The sensitivity and specificity of ctDNA detectionUnknown
NCT05208372, ChinaProspective case–control;
Biomarkers:
200Value of CTCs and ctDNA in the diagnosis of metastasis in ascites/peritoneal flushing fluid and bloodQuantity of CTCs; Expression of ctDNAUnknown
NCT04576858, DenmarkProspective cohort;
Biomarkers: ctDNA
1950To evaluate the treatment effect as well as predictive and prognostic factors with special emphasis on the clinical utility of ctDNA in plasma in patients with GE cancerTime to recurrenceUnknown
NCT05348161, ChinaInterventional non-randomized parallel:
Biomarkers: HER2/PD-L1-positive CTCs; ctDNA genomic events
100To evaluate how HER2-targeted therapy and immunotherapy affect molecular profiles in HER2-positive gastric cancer patients via multi-omics liquid biopsy markersProportions of HER2- and PD-L1-positive CTCs
Incidence rates of various ctDNA genomic alterations (e.g., copy number changes, insertions/deletions)
Unknown
NCT07076979, ChinaProspective case–control;
Biomarkers: Metabolic markers
250To develop and validate metabolic biomarkers for early diagnosis, prognosis, and prediction of recurrence and metastasis in gastric cancerOS, DFS, HR, PPV and NPV Ongoing, recruiting
NCT04000425, ChinaProspective cohort;
Biomarkers: AVENIO ctDNA surveillance kit
55Evaluate ctDNA as an indicator of MRD and as a marker of adjuvant chemotherapy response after radical gastrectomy.Disease recurrence risk; DFS; ctDNA changing to adjuvant chemotherapy response; Time of first negative ctDNA detection from positive ctDNA detectionUnknown
NCT05366881, United StatesProspective multicenter case–control;
Biomarkers: Genome-wide cfDNA methylome enrichment
7000Train and validate methylation-based ctDNA test for early cancer detection and MRD (including esophagus and gastric)Sensitivity & specificity of assay vs. controlsOngoing, recruiting
NCT07035587, KoreaProspective-retrospective cohort;
Biomarkers: Serial cfDNA (ctDNA), RNA, protein profiles
1200Early diagnosis & post-treatment MRD monitoring for multiple cancers (including esophagus and gastric)Sensitivity & specificity for cancer detection; VAF correlation with recurrenceOngoing, recruiting
NCT05059444, United States, Germany, France, Italy and SpainProspective multicenter cohort;
Biomarkers: Guardant reveal assay using methylated ctDNA
2020Using a novel ctDNA approach to detect recurrence in early-stage solid tumors (including esophagus and gastric cancer)Distant recurrence free interval. Lead time, sensitivity, and specificity and of ctDNA in detecting recurrence Ongoing, recruiting
NCT06227728, VietnamProspective multicenter cohort; Biomarkers: ctDNA using targeted sequencing and multiplex PCR approaches.50Assess if changes in ctDNA can predict early response to ICIs in patients with advanced-stage cancer (including gastric cancer)Association between ctDNA dynamics and clinical response; comparison with RECIST; prognostic value of ctDNA clearance and with PFS/OS Ongoing, recruiting
NCT04168931, BrazilInterventional Phase II open label;
Biomarkers: HER-2 positive CTCs
85To investigate whether HER2-expressing CTCs may be suitable for prediction of response in patients with relapsed or metastatic gastric cancer who are histologically HER2-negative and treated with trastuzumab combination chemotherapyRadiological response rate, frequency of HER 2 expression among CTCs of patients with recurrence or metastasis with negative expression in tumor tissueTerminated, recruitment failure
NCT03023436, ChinaInterventional Phase III open label trial;
Biomarkers: ctDNA and CTCs
220To assess ctDNA and CTC alterations as potential biomarkers for debulking surgery combined with HIPEC and systemic chemotherapy in patients with gastric cancer and peritoneal dissemination (as a secondary outcome measure)Median survival time, OS, PFS, morbidity and mortality, QOL, CTCs changes, ctDNA changes, and molecular biomarker (including 14 genes) alterationUnknown status
NCT04510285, United StatesInterventional Phase II open label;
Biomarkers: ctDNA
48To evaluate differences in 6-month ctDNA clearance rate in HER2+ esophagogastric cancer with persistent ctDNA
following curative surgery when treated with “second
adjuvant” trastuzumab with or without pembrolizumab
Rate of ctDNA clearance at 6 months Terminated, recruitment failure
NCT04665687, Korea Prospective cohort; Biomarkers: ctDNA1730To differentiate early gastric cancer and precancerous adenoma and predict recurrence by finding biomarkers through molecular profilingBiomarker-based differentiation between adenoma and early GC; prognostic biomarkers for recurrenceUnknown status
NCT05594381, ChinaInterventional Phase II open label;
Biomarkers: ctDNA
90To dynamically detect gene mutations, protein expressions and tumor images in G/GEJ tumor tissues and blood samples before, under and after PD-1 monoclonal antibody (sintilimab) combined with SOX neoadjuvant therapy by using ctDNA targeted sequencing combined with multi-omics technologypCR, ORR, DCR, MPR, TRG, R0 resection rate, OS, tumor downstaging, DFS, treatment-emergent adverse events and 30-day postoperative mortalityNot yet recruiting
NCT04929015, United StatesInterventional open label;
Biomarkers: tumor-informed personalized ctDNA assay (Signatera)
30Utility of ctDNA as a sensitive biomarker in patients with Peritoneal Carcinomatosis treated with chemotherapy, CRS and/or HIPECClearance rate of ctDNA with cytoreductive surgery (CRS), comparing with clinical staging of CRS and activity of chemotherapy in this diseaseOngoing, recruiting
NCT05482516, United StatesInterventional Phase III open label;
Biomarkers: tumor-informed personalized ctDNA-MRD assay (Signatera)
20Guide atezolizumab and bevacizumab therapy by MRD status in GI cancerRate of Signatera ctDNA positivity, rate of enrollment, rate of ctDNA complete response, rate if ctDNA partial disease and rate of ctDNA progressive diseaseOngoing, recruiting
NCT05661110, ChinaProspective cohort;
Biomarkers: AmoyDx® Master Panel (559 genes for DNA mutation and 1813 genes for RNA expression)
46To analyze the correlation between genomic alterations, gene expression characteristics and the efficacy of HIPEC combined with PD1/PDL1 inhibitor conversion therapy in patients with peritoneal metastasis of gastric cancerRelationship between the status, numerical changes in ctDNA during HIPEC combined with PD1/PDL1 inhibitor conversion therapy and postoperative R0 resection rate. Correlation between genomic changes of ctDNA and ORR, OS, RFA and event-free survivalNot yet recruiting
NCT04943406, ItalyProspective cohort;
Biomarkers: ctDNA
150Prognostic role of ctDNA in patients with locally advanced gastric cancerImpact of ctDNA (in peritoneal lavage and peripheral blood) positivity on OS and DFSOngoing, recruiting
NCT06253650, ItalyInterventional phase II open label;
Biomarkers: ctDNA
46To investigate the activity, efficacy and safety of trastuzumab-deruxtecan (T-DXd) plus capecitabine/5-fluorouracil as a postoperative treatment in localized/locally advanced gastric or GE junction cancer (GC/GEJC)/esophageal adenocarcinoma patients with HER2 overexpression/amplification and positive postoperative ctDNA after preoperative 5-fluorouracil plus leucovorin, oxaliplatin, and docetaxel (FLOT) regimen followed by radical surgeryctDNA clearance, DFS, OS, metastases-free survival, and QOLOngoing, recruiting
NCT05494060, ChinaInterventional Phase II open label;
Biomarkers: ctDNA
80To assess safety and anti-tumor activity Penpulimab in combination with Anlotinib and standard chemotherapy as adjuvant treatment for ctDNA-positive G/GEJ cancerDFS at different time points, OS and toxicity Ongoing, recruiting
NCT05965479, United KingdomInterventional Phase II open label;
Biomarkers: ctDNA (Signatera assay)
25To assess the efficacy of trastuzumab deruxtecan in reducing micrometastatic disease burden in HER2 positive GEA patients who are ctDNA positive after chemotherapy and surgeryctDNA clearance, DFS, OS, and QOLOngoing, recruiting
NCT05067842, United StatesProspective case–control;
Biomarkers: ctDNA (Signatera assay)
30To determine the feasibility of assessing tumor response utilizing ctDNA in patients of locally advanced esophageal and GE junction (LA-EA/GEJ) cancerTumor response measured by ctDNA, R0 surgical resection, OS and RFSWithdrawn
NCT06498752, ChinaInterventional Phase II open label;
Biomarkers: ctDNA
102To validate whether ctDNA MRD status after radical radiotherapy can stratify prognosis and guide consolidation therapy with PD-1 inhibitors in patients with ESCCMedian PFS, OS, cancer-specific survival, toxicity, swallowing function, dynamic ctDNA changes and correlation with recurrenceOngoing, recruiting
NCT05759325, China Prospective cohort;
Biomarkers: ctDNA MRD
100To observe and evaluate the correlation between ctDNA-MRD and the therapeutic effect and prognosis of stage II-IVA operable ESCCPFS rate of ESCC patients with different MRD status during perioperative periodNot yet recruiting
NCT06103890, ChinaProspective cohort;
Biomarkers: ctDNA
100To explore the clinical value of MRD as a biomarker for assessing treatment efficacy, predicting recurrence risk, and evaluating prognosis in ESCCpCR, R0 resection rate, ctDNA clearance, MPR, RFS, and OSOngoing, recruiting
NCT05426850, ChinaProspective cohort;
Biomarkers: ctDNA
100To analyze the relationship between the dynamic changes in ctDNA and tumor relapse of ESCC treated by concurrent chemoradiotherapyChanges in ctDNA status and recurrence, OS, and RFS. Unknown status
Abbreviation: NCT: 2. Human Epidermal growth factor Receptor 2, HIPEC: hyperthermic intraperitoneal chemotherapy, PD1: programmed cell death protein 1, PDL1: programmed death-ligand 1, ORR: objective response rate, DCR: disease control rate, MPR: major pathological response, TRG: tumor regression grade, DFS: disease-free survival, QOL: quality of life, G/GEJ: gastric/gastroesophageal junction, ICI: immune checkpoint inhibitor, ESCC: esophageal squamous cell carcinoma, VAF: variant allele frequency, CRS: cytoreductive surgery.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dadgar, N.; Anees, M.; Sherry, C.; Park, H.Y.; Grayhack, E.E.; Goel, A.; Khan, A.F.; Omstead, A.; Bartlett, D.L.; Wagner, P.L.; et al. Blood-Based Surveillance Biomarkers for Gastroesophageal Cancers. Cancers 2025, 17, 3552. https://doi.org/10.3390/cancers17213552

AMA Style

Dadgar N, Anees M, Sherry C, Park HY, Grayhack EE, Goel A, Khan AF, Omstead A, Bartlett DL, Wagner PL, et al. Blood-Based Surveillance Biomarkers for Gastroesophageal Cancers. Cancers. 2025; 17(21):3552. https://doi.org/10.3390/cancers17213552

Chicago/Turabian Style

Dadgar, Neda, Muhammad Anees, Christopher Sherry, Hyun Young Park, Erin E. Grayhack, Arul Goel, Alisha F. Khan, Ashten Omstead, David L. Bartlett, Patrick L. Wagner, and et al. 2025. "Blood-Based Surveillance Biomarkers for Gastroesophageal Cancers" Cancers 17, no. 21: 3552. https://doi.org/10.3390/cancers17213552

APA Style

Dadgar, N., Anees, M., Sherry, C., Park, H. Y., Grayhack, E. E., Goel, A., Khan, A. F., Omstead, A., Bartlett, D. L., Wagner, P. L., & Zaidi, A. H. (2025). Blood-Based Surveillance Biomarkers for Gastroesophageal Cancers. Cancers, 17(21), 3552. https://doi.org/10.3390/cancers17213552

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop