Open Access

Cell‑free DNA as a liquid biopsy for early detection of gastric cancer (Review)

  • Authors:
    • Zheng-Bin Huang
    • Hai-Tao Zhang
    • Benjamin Yu
    • De-Hua Yu
  • View Affiliations

  • Published online on: November 3, 2020     https://doi.org/10.3892/ol.2020.12264
  • Article Number: 3
  • Copyright: © Huang 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

Gastric cancer (GC) is one of the most common malignant tumors with poor prognosis worldwide, mainly due to the lack of suitable modalities for population‑based screening and early detection of this disease. Therefore, novel and less invasive tests with improved clinical utility are urgently required. The remarkable advances in genomics and proteomics, along with emerging new technologies for highly sensitive detection of genetic alterations, have shown the potential to map the genomic makeup of a tumor in liquid biopsies, in order to assist with early detection and clinical management. The present review summarize the current status in the identification and development of cell‑free DNA (cfDNA)‑based biomarkers in GC, and also discusses their potential utility and the technical challenges in developing practical cfDNA‑based liquid biopsy for early detection of GC.

Introduction

Despite significant progress being made in the prevention and treatment of gastric cancer (GC) in the past decades, GC is still one of the most concerning malignancies as the majority of patients are diagnosed at an advanced stage of disease. Globally, GC ranked fifth for cancer incidence and third for cancer deaths, accounting for 1.3 million estimated incident cases and 819,000 estimated deaths in 2015 (1). Geographically, GC is more prevalent in developing countries, with the majority of cases and deaths occurring in Eastern Asia, including China, Japan and Korea, followed by Central Europe, Eastern Europe and South America (2). Etiologically, GC is a multifactorial disease attributed to both host and environmental factors. Proposed risk factors for GC include Helicobacter pylori (H. pylori) infection, smoking, alcohol, obesity, salt intake, atrophic gastritis (AG), intestinal metaplasia (IM) and family history of GC. The current therapy for GC includes surgery, chemotherapy, radiotherapy, VEGFR (ramucirumab) and targeted therapy against HER2 (trastuzmab) (3). The overall outcome of GC is largely associated with the stage of the disease at diagnosis. For early GC limited to the mucosa and submucosa, the 5-year survival rate is >90% (4,5). However, due to the lack of distinguishable symptoms at early stages and effective mass screening programs worldwide, the majority of GC cases are typically detected at stage IIIA-IV, with an estimated 5-year survival rate of <30% and a median survival of 12 months (4,5).

To date, four GC screening methodologies have been implemented in clinical settings: H. pylori serology, serum pepsinogen (PG) testing, indirect upper gastrointestinal series (UGIS) and endoscopy. Since the 1960s, population-based GC screening programs in several high-prevalence nations such as Japan and Korea have achieved significantly improved survival and cure rates using the above methods. These programs demonstrate the effectiveness of mass screening for GC (68). However, each of these screening tools has its limitations. For instance, H. pylori serology is unable to detect premalignant lesions, such as longstanding AG and IM. Therefore, H. pylori serology alone is not useful as a screening test for GC (9). The combination of upper endoscopy with pathological biopsy examination is the primary screening technique in the majority of these programs and the gold standard for confirmation of diagnosis (10). In general, endoscopy is superior to UGIS in sensitivity and cost-effectiveness for detecting early GC (1113). However, endoscopy is an invasive technique that has infrequent but serious complications, and its utility depends largely on the skill of the endoscopist (14). Therefore, the use of endoscopy in mass-screening programs in low-prevalence and low-income countries is impractical and likely to be associated with low participation rates. Currently, the only non-invasive test for GC detection in the clinical setting is the PG assay (15). Changes in serum PG levels reflect the function of the gastric mucosa. Decreased PGI levels and PGI/PGII ratio are indicators of atrophic changes in the gastric corpus. PG tests can detect gastric mucosal atrophy with a sensitivity of 66.7–84.6% and a specificity of 73.5–87.1% (16,17). However, PG assay's sensitivity for GC detection ranges from 36.8 to 62.3% (18,19), which is too low to be acceptable for population-based screening. Therefore, new assays with improved sensitivity, specificity and cost-effectiveness are needed.

Recent advances in genetic testing, such as next-generation sequencing (NGS) and digital PCR, and bioinformatics have accelerated the research on liquid biopsy greatly, which have high potential to change the clinical management of patients with cancer. Meanwhile, considerable efforts have been made to identify novel, early-stage GC biomarkers with potential utility in clinical liquid biopsy testing. Such biomarkers include cell-free DNA (cfDNA), cell-free RNA (cfRNA), proteins, autoantibodies, circulating tumor cells (CTCs), cancer-derived extracellular vesicles (EVs) and metabolites. A number of comprehensive reviews were recently published on CTCs, proteomics, cfRNA biomarkers, exosomes and EVs in GC (20,21). The current review provides an overview of the recent advances in the early detection of GC using liquid biopsy, with a focus on cfDNA, and their origin and mechanism of release into the bloodstream, as well as their potential utility in clinical practice (Fig. 1).

Quantification of circulating cfDNA

cfDNA is circulating extracellular DNA existing in the blood serum or plasma, synovial fluid, cerebrospinal fluid and other body fluids. Physiological events producing cfDNA include cellular apoptosis, secretion, micrometastasis and necroptosis (22). In patients with cancer, the plasma cfDNA levels are 2–3-fold higher than those in normal healthy groups (23), suggesting its potential as a complementary biomarker for cancer detection, as well as an indicator of prognosis and therapeutic response. As shown in Table I, a prospective study quantified the cfDNA levels in serum samples from patients with benign or malignant gastrointestinal tract disease using a radioimmunoassay, and it showed that the cfDNA levels in patients with malignant diseases were significantly higher than those of patients with benign diseases (24). Consistently, Sai et al (25) reported that increased plasma total cfDNA in patients with GC could be detected compared with the undetectable levels of healthy controls (Table I). The authors measured the short and long forms of β-actin in plasma samples from patients with GC and healthy controls by quantitative PCR (qPCR), and found that the cfDNA concentration in patients with GC was significantly higher. These studies suggest the potential use of serum cfDNA concentration assay to detect GC.

Table I.

cfDNA as biomarker for detection of gastric cancer.

Table I.

cfDNA as biomarker for detection of gastric cancer.

Diagnostic value

cfDNABiomarker candidateSample typeMethodSensitivitySpecificity(Refs.)
cfDNAcfDNA levelsSerumRadioimmuno assayMean concentration of 412 and 118 ng/ml respectively (P<0.01) (24)
PlasmaqPCRBy 102 bp β-actin assay, GC=5.71 ng/ml, HC=3.20 ng/ml (P=0.03) By 253 bp β-actin assay, GC=0.470 ng/ml, HC=0.212 ng/ml (P<0.0001) (25)
PlasmaAlu81-qPCR75% (41/54)  63% (37/59)(26)
PlasmaMeasurement of cfDNA concentration96.67% (29/30)94.11% (32/34)(27)

[i] cfDNA, cell-free DNA; qPCR, quantitative PCR; bp, base pair.

There are various methods for quantitative cfDNA detection, but their efficiency is limited by sample preparation and assay procedures. To address this issue, Park et al (26) developed an Alu-qPCR assay for measuring cfDNA concentrations, which demonstrated better sensitivity and reproducibility compares with other technologies, based on Ultraviolet-visible (UV–Vis) spectrophotometry or the PicoGreen fluorophore. Applying this assay, the cfDNA levels were compared between patients with GC and those of age-matched healthy controls, and found that the mean levels of plasma cfDNA were higher in the GC group than in the control group (Table I). To understand the dynamics of cfDNA levels pre- and post-surgery, Kim et al (27) measured the plasma cfDNA levels of patients with GC and healthy controls by qPCR. The samples of the patients with GC were collected before and 24 h after surgery. The results showed that average cfDNA levels were increased in patients with GC compared with those of healthy controls, and there was a positive dose-dependent association with more advanced cancer staging. Meanwhile, the levels of cfDNA in the 24-h-post-surgery group decreased significantly, thus supporting the utility of using cfDNA to monitor disease severity and therapeutic efficacy.

Recently, Qian et al (28) investigated cfDNA levels in serum samples from 124 patients with GC, 64 patients with benign gastric disease (BGD; namely gastric adenoma) and 92 healthy controls using the Alu-qPCR assay. The results showed that cfDNA levels were significantly higher in patients with GC compared with those with BGD or in healthy controls (P<0.05). Further statistical analysis showed that serum cfDNA levels in the GC group were significantly associated with advanced staging (III–IV) and tumor size (>5 cm), but not with sex, age or tumor location. cfDNA was also more sensitive in the detection of stage-I GC than conventional tumor biomarkers, including carcinoembryonic antigen (CEA), carbohydrate antigen (CA)-19-9, CA50 and CA72-4, suggesting that cfDNA assays may be able to replace current protein tumor biomarkers for cancer detection.

Since the measurement of cfDNA levels does not require any prior knowledge of genetic alterations in tumor tissue, this test could be highly useful in non-invasive assays for early GC detection. However, the application of cfDNA quantification alone for early cancer diagnosis is limited by several obstacles: i) Circulating cfDNA is unstable and its kinetics has not yet been well defined, which may affect assay robustness and standardization; ii) cfDNA levels cannot differentiate cancer type or tissue of origin; iii) cfDNA testing is relatively nonspecific, as numerous patients with non-cancer conditions, such as inflammatory disease, infections and cardiovascular disease, and even healthy individuals after exercise, show elevated cfDNA levels (29,30). Therefore, it is expected that the combined detection of cfDNA levels with other markers may achieve improved clinical performance.

Detection of genetic alteration of circulating tumor DNA (ctDNA)

Genetic alterations such as mutations, rearrangement and amplification of driver genes result in tumorigenesis. Recent advancements in NGS techniques have identified various nucleotide mutations associated with GC (Table II). The top mutated genes showing higher mutation frequency were TP53, TTN, MUC16, CDH1, KMT2C and MLH1 (3133). In 1977, Leon et al (34) reported that numerous patients with cancer had elevated circulating cfDNA, and found that this DNA was tumor derived. Since, scientists focused their attention on the content of ctDNA. Some of the aberrant DNA shed into the blood by cancer cells, such as EGFR and KRAS mutations, can be potential biomarkers. Previous studies have demonstrated the detectability of mutant DNA released from tumor tissues. In fact, ctDNA analysis has emerged as an additional diagnostic tool to guide clinical management of certain cancer types including lung cancer and colon cancer (35,36). However, the use of ctDNA for early cancer detection is complicated by the generally low abundance of ctDNA in early-stage cancer and the technical challenges in its detection.

Table II.

Type and frequency of mutations implicated in gastric cancer.

Table II.

Type and frequency of mutations implicated in gastric cancer.

No. of patientsMethodTop mutated genesMutation frequency (%)(Refs.)
74WESTP5348(31)
TTN37
MUC1615
ABCA1312
SYNE112
DCHS211
HMCN111
OBSCN11
ROBO111
63WESTP5348(32)
TTN37
MUC1614
ABCA1313
DCHS213
DNAH1111
HMCN111
LAMA111
PCLO11
ROBO111
SYNE111
43WESTP5341(33)
CDH126
KMT2C24
MLH118
SMAD415
GNAS15
CDKN2A12
RPL512
TAF112
SETD212
PTEN12

[i] WES, whole-exome sequencing; TP53, tumor protein p53; TTN, Titin; mucin 16, cell surface associated; ABCA13, ATP binding cassette subfamily A member 13; DCHS2, dachsous cadherin-related 2; SYNE1, spectrin repeat containing nuclear envelope protein 1; HMCN1, hemicentin 1; OBSCN, obscurin, cytoskeletal calmodulin and titin-interacting RhoGEF; ROBO1, roundabout guidance receptor 1; LAMA1, laminin subunit alpha 1; PCLO, piccolo presynaptic cytomatrix protein; CDH1, cadherin 1; KMT2C, lysine methyltransferase 2C; MLH1, mutL homolog 1; SMAD4, SMAD family member 4; GNAS, GNAS complex locus; CDKN2A, cyclin dependent kinase inhibitor 2A; RPL5, ribosomal protein L5; TAF1, TATA-box binding protein associated factor 1; SETD2, SET domain containing 2, histone lysine methyltransferase; PTEN, phosphatase and tensin homolog.

Applying the droplet digital PCR (ddPCR) assay, which is currently the most sensitive method, Bettegowda et al (37) evaluated ctDNA in 640 plasma samples from patients with various cancer types and stages. ctDNA was detected in >75% of patients with several advanced cancer types, including melanoma, and pancreatic, breast, hepatocellular, ovarian, colorectal, bladder, gastroesophageal, and head and neck cancer. In patients with localized tumors, ctDNA was detectable only in a subset of patients with gastroesophageal cancer (57%), colorectal cancer (CRC) (73%), pancreatic cancer (48%) and breast adenocarcinoma (50%). When KRAS mutations were tested for in ctDNA in an additional panel of 206 patients with metastatic CRC, the sensitivity and specificity of detection were 87.2 and 99.2%, respectively. These results indicate that the detectability of ctDNA is affected by cancer type and stage.

Recently, Cohen et al (38) developed a blood-based test (CancerSEEK) and investigated its utility in the early detection of eight common cancer types, such as ovarian, liver, stomach, pancreatic, esophageal, colorectal, lung and breast cancer. The above blood test couples targeted parallel sequencing of ctDNA with eight known protein biomarkers using a panel of 61 amplicons in 16 genes, including TP53, KRAS, PIK3CA, CTNNB1 and APC. The authors applied this assay to 1,005 patients with clinically detected stage I–III cancer, and the method yielded 70% of median positivity for all eight cancer types tested and >99% specificity for healthy controls. Using this assay, the authors were also able to track the cancer origin to two possible sites in ~80% of patients. As shown in Table III, GC samples were included in this study, and mutations of a number of driver genes were detected. This study suggests the divergent utility of a single test in detecting multiple early-stage cancer types.

Table III.

ctDNA as biomarker for detection of gastric cancer.

Table III.

ctDNA as biomarker for detection of gastric cancer.

Diagnostic value, % (n)

TypeBiomarker candidateSample typeMethodSensitivitySpecificity(Refs.)
ctDNAGene mutations (TP53, KRAS, PIK3CA, CTNNB1, APC, PPP2R1APlasmaNGS72 (49/68)  99 (805/812)(38)
MYC gene copy numberPlasma   qPCR75.4 (43/57)76.9 (60/79)(41)
(MYC/GAPDH ratio and MYC/HBB ratios)PlasmaqPCR72.8 (59/81) for MYC>2.72577.7 (80/103)(42)
HER2 and MYC gene copy number (HER2/HBB and MYC/HBB ratios)PlasmaqPCR69.1 (56/81) for both HER2>2.0 and MYC>2.72592.2 (95/103)
HER2 gene copy numberPlasmaqPCR87.7 (71/81) for HER2>2.064.1 (66/103)
Tissue and serumddPCR29.2 (7/24) (43)
PlasmaqPCRFor discovery: 53.9 (7/13) For validation: 66.7 (2/3)For discovery: 96.7 (29/30) For validation: 100 (22/22)(44)

[i] TP53, tumor protein p53; KRAS, kirsten rat sarcoma viral oncogene; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha; CTNNB1, catenin beta-1gene; APC, adenomatous polyposis coli gene; PPP2R1A, protein phosphatase 2 regulatory subunit α; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; HBB, hemoglobin beta; HER2, human epidermal growth factor receptor 2; NGS, Next generation sequencing; ddPCR, digital droplet PCR.

In addition to mutations, the copy number variants of numerous genes such as c-MYC and HER2 were also identified in GC by genome-wide profiling (39,40). Applying a qPCR assay, Park et al (41) measured the MYC/GAPDH ratio in plasma samples from patients with GC and cancer-free individuals. This showed that the mean ratio of MYC/GAPDH in plasma was significantly increased in GC compared with that in healthy controls (P<0.001). Another study compared the plasma levels of HER2 and MYC genes in patients with GC, gastric adenoma, gastritis or no disease with those in matched tissue samples by reverse transcription-qPCR (42). The results indicated that the HER2/HBB and MYC/HBB ratios in tissue and plasma from patients with GC were significantly increased compared with those in gastritis tissue and cancer-free individuals. Similarly, Kinugasa et al (43) detected increased HER2 in ctDNA from serum samples in patients with GC by ddPCR. Recently, Shoda et al (44) investigated the HER2 gene levels in plasma samples of patients with GC and healthy controls using ddPCR. The results showed that the preoperative plasma HER2 ratio (normalized with an internal control) correlated with HER2-positivity status in the tumor (P<0.001) (Table III). Of note, although the increased levels of circulating HER2 and c-MYC genes were detectable in GC, their potential utility in early diagnosis would be limited by their low positivity rates in patients with GC.

Measurement of cfDNA methylation

DNA modifications such as 5-methylcytosine (5-mC) and 5-hydroxymethylcytosine (5-hmC) could serve as ideal biomarkers for cancer diagnosis. The 5-mC remodeling of DNA has been reported to be involved in cancer initiation, progression and therapeutic response (45). Except for 5-mC, a previous study by Li et al (46) showed that 5-hmC from circulating cfDNA was highly predictive of colorectal and gastric cancer, and was superior to conventional biomarkers and comparable to 5-hmC biomarkers from tissue biopsies. Hypermethylation of promoter CpG islands in tumor suppressor genes plays a crucial role in carcinogenesis (4749). Eyvazi et al (50) verified the promoter methylation of EphA5, HS3ST2 and CDH11 genes in patients with GC using paraffin-embedded tissue sections. Recently, cfDNA in blood plasma has become a promising cancer biomarker for early diagnosis (51). The abnormal methylation of a large number of genes has been demonstrated to have utility for non-invasive detection of cancer in plasma or serum samples (5254). Among them, Septin 9 gene methylation, detectable as hypermethylated Septin 9 DNA fragments in blood plasma, is a front-runner for the clinical screening of CRC (55). Septin 9, a member of the Septin family, was originally identified in myeloid neoplasia (56). It functions as a tumor suppressor gene in multiple cancer types (57). Consequently, several assay kits have been developed to detect methylated Septin 9. Epi proColon, the first commercial methylated Septin 9 assay, has been approved by the USA Food and Drug Administration (FDA) for average-risk patients over the age of 50 years, and has also been approved in Europe and China (58). Meta-analysis of existing clinical data showed that the assay's sensitivity and specificity were 71 and 92%, respectively, demonstrating its reliability for CRC detection (59,60). In 2016, a blood test that detects circulating methylated Septin 9 DNA was approved by the FDA to provide an alternative screening modality (61), thus paving the way for the development of a new generation of liquid biopsy tests for early cancer detection.

Besides Septin 9, there is increasing evidence demonstrating that many other hypermethylated genes, including Reprimo, Rassf1A, CDH1, CDKN2A, MLH1, RUNX3, APC and p16, can be differentially detected in plasma or serum samples of patients with GC (Table IV). Among them, p16 gene hypermethylation in GC has been extensively studied. p16 is a cell-cycle regulator that arrests cells in G1 phase by inhibiting cyclin D-dependent protein kinase (CDK)4 and CDK6 (62). It functions as a tumor suppressor and can be inactivated by hypermethylation in the gene-promoter region (63). Kanyama et al (64) investigated the promoter methylation status of the p16 gene in paired tumor and serum samples from patients with GC. p16 hypermethylation was found in primary GC samples but in none of the corresponding gastric mucosae. Consistently, Ichikawa et al (65) reported that hypermethylation of the promoter region in the p16 and E-cadherin genes was detected in serum DNA samples from patients with GC, but not from healthy volunteers. Recently, Guo et al (66) detected promoter methylation of the p16 gene in peripheral blood samples from patients with GC and healthy controls using methylation-specific PCR (MSP) analysis, and found that the ratio of methylated p16 was significantly higher in GC samples compared with the control group (P<0.01). However, the reported ratios of circulating p16 methylation in GC vary remarkably across different studies (67). Thus, further validations are needed to determine whether the variation in positive methylation ratios is due to assay aberrations or primary sample differences.

Table IV.

Methylated DNA as biomarker for detection of gastric cancer.

Table IV.

Methylated DNA as biomarker for detection of gastric cancer.

Diagnostic value, % (n)

cfDNABiomarker candidateSample typeMethodSensitivitySpecificity(Refs.)
MethyDNAP16SerumMSP  26 (6/23)  100 (16/16)(64)
SerumMSP  18 (20/109)  100 (10/10)(65)
BloodMSP  72.6 (77/106)94.4 (17/18)(93)
SerumMSP51.9 (28/54)  100 (30/30)(98)
PlasmaMSP/HPLC14.3 (12/84)  100 (15/15)(110)
SerumMSP26.9 (14/52)  100 (29/29)(67)
SerumMSP  22 (9/41)  100 (10/10)(111)
P16/E-CadherinSerumMSP  37 (40/109)  100 (10/10)(65)
E-CadherinSerumMSP  24 (26/109)  100 (10/10)
SerumMSP  22 (9/41)  100 (10/10)(111)
p16/E-cadherin/RARbetaSerumMSP  44 (18/41)  100 (10/10)
RUNX3Tissues and cell linesMSP  64 (48/75) (69)
Serumrt-MSP70.8 (143/202)  99.8 (848/850)(71)
SerumqMSP95.562.5(70)
PlasmaMSP  42.7 (56/131)  100 (34/34)(72)
Plasma ZIC1, HOXD10 and RUNX3PlasmaMSP91.650.0
Zic1PlasmaMSP  69.5 (91/131)  100 (34/34)
RASSF1ASerumMSP  34 (16/47)  100 (30/30)(74)
SerumMSP68.5 (50/73)  100 (20/20)(75)
PlasmaMSP  83.2 (84/101)  94.55 (191/202)(76)
PCDH10PlasmaMSP  94.1 (95/101)  97.03 (142/202)
ReprimoPlasmaMSP95.3 (41/43)90.3 (28/31)(79)
PlasmaMSP86.3 (44/51)97.6 (48/49)(81)
PlasmaMSP  62 (31/50)  100 (30/30)(80)
hMLH1PlasmaMSP  48 (24/50)96.7 (29/30)

[i] P16, Cyclin-dependent kinase inhibitor 2A, multiple tumor suppressor 1; E-Cadherin, epithelial cadherin; RARbeta, retinoic acid receptor beta; RUNX3, Runt-related transcription factor 3; ZIC1, zinc finger of the cerebellum 1; RASSF1A, Ras association domain-containing protein 1; PCDH10, protocadherin-10 gene; hMLH1, MutL homolog 1; MSP, methylation-specific polymerase chain reaction; HPLC, high performance liquid chromatography.

Besides p16, Runt-related transcription factor 3 (RUNX3) has also been reported as a candidate tumor suppressor in GC (68). Hypermethylation of RUNX3 CpG islands was observed in GC cell lines and primary gastric carcinoma, with significantly higher ratios than in non-malignant gastric disorders (69). Consistently, high levels of methylated RUNX3 sequences were also detected in the peripheral circulation of patients with GC. The RUNX3 methylation index was concordant with cancer stage, histology, and lymphatic and vascular invasion, and was more sensitive than CEA as a biomarker (70). Applying a real-time MSP assay, Lu et al investigated RUNX3 methylation levels in serum samples from normal individuals without any gastric lesions or H. pylori infection, patients with benign lesions and patients with GC (71). Notably, the serum detection of RUNX3 methylation was negative in almost all benign and normal samples, except for two patients who had severe dysplasia. However, circulating methylated RUNX3 was detected in almost all patients with GC who had detectable RUNX3 methylation in tissue samples with significant accordance (k=0.887; P<0.001) (71). Recently, Lin et al (72) measured the methylation status of three selected genes in blood samples of patients with GC and precancerous lesions using an MSP assay, and found that the methylation rate of ZIC1, HOXD10 and RUNX3 increased significantly during the progression of gastric carcinogenesis. For predicting GC and intraepithelial neoplasia, the combined detection of these three genes showed a synergistic effect compared with that of testing a single biomarker.

Ras-association domain family 1A (RASSF1A) is a putative tumor suppressor gene, which is often silenced by hypermethylation of its promoter region in a number of human tumors such as GC (73). Previous studies conducted by Wang et al (74) and Balgkouranidou et al (75) reported that RASSF1A methylation was detected in the serum samples of patients with GC, but not in the healthy control samples (P<0.001). Recently, Pimson et al (76) investigated the promoter methylation statuses of RASSF1A and PCDH10 (another tumor suppressor gene in the protocadherin family) in GC. The authors found that hypermethylation of PCDH10 and RASS1A was detectable in plasma samples from patients with GC, and aberrant PCDH10 and RASSF1A promoter methylation in plasma DNA was associated with worse clinical outcome.

Reprimo (RPRM) is another tumor suppressor gene involved in the development of numerous malignant tumors, including GC (77,78). Bernal et al (79) evaluated the DNA methylation patterns of 24 genes by MSP in primary tissues from patients with GC. In >50% of cases, hypermethylation was detected in ≥1 of 11 genes, including APC, SHP1, E-cadherin, ER, Reprimo, SEMA3B, 3OST2, p14, p15, DAPK and p16. The most frequently hypermethylated genes were further evaluated in primary tissues and plasma samples from prospectively collected GC cases, as well as in plasma samples from asymptomatic age- and gender-matched controls (which formed the validation group). The results confirmed a high methylation frequency of seven genes (namely, APC, SHP1, E-cadherin, ER, Reprimo, SEMA3B and 3OST2) in GC. Notably, the prevalence of Reprimo methylation was significantly higher in GC tumor and plasma samples than in asymptomatic control samples (P<0.001). In another study (80), the DNA methylation statuses of the Reprimo and hMLH1 genes in tissue and plasma samples of patients with GC, dysplasia, chronic AG and normal controls were investigated by MSP. The results showed that plasma hypermethylation of Reprimo was detectable in GC, AG and dysplasia, but not in normal controls. Meanwhile, methylated hMLH was also detected in a higher percentage of GC and dysplasia samples compared with that of normal controls. Consistently, Lai et al (81) examined Reprimo gene methylation in GC tissues and plasma samples by bisulfite sequencing, and found that the Reprimo gene-promoter region was hypermethylated in GC tissues, plasma and cell samples. This was correlated with a decrease in Reprimo gene expression, thus supporting the potential utility of Reprimo methylation as a diagnostic biomarker for GC.

Of note, there is obvious heterogeneity in gene methylation frequencies between different studies, which might be attributable to differences in samples size, technique variations and geographical differences. To obtain a better understanding of this variance, Hu et al (82) recently performed a meta-analysis to evaluate the pooled sensitivity and specificity of the results of 32 studies, which included 4,172 patients with GC and 2,098 controls. Collectively, the overall sensitivity of DNA methylation-based blood test for detecting GC was 57% (95% CI, 50–63%), while the specificity was 97% (95% CI, 95–98%). The sensitivity and specificity of tests covering multiple methylated genes were 76% (95% CI, 64–84%) and 85% (95% CI, 65–95%), respectively. These results indicate that blood-based DNA methylation tests have high specificity but modest sensitivity for detecting GC. Evaluating multiple methylated genes or using plasma samples seems to improve diagnostic sensitivity.

Besides the methylation markers described above, increased methylation of numerous other genes in cfDNA has also been reported in GC (8386) (Table V). However, due to the limited sample sizes and method variants, further studies are needed to demonstrate their analytical and clinical validity.

Table V.

Methyated genes with limited sample sizes and method variants.

Table V.

Methyated genes with limited sample sizes and method variants.

Diagnostic value, % (n)

TypeBiomarker candidateSample typeMethodSensitivitySpecificity(Refs.)
MethyDNAZic1PlasmaMSP  60.6 (63/104)  100 (20/20)(83)
RASSF10SerumBSP  81.71 (67/82)  89.5 (85/95)(84)
RNF180Plasmaq-MSP  56 (18/32)  100 (64/64)(85)
SFRP1SerumMSP  30.95 (13/42)  93.2 (41/44)(86)
IRX1PlasmaMSP  73.3 (11/15)  90 (9/10)(112)
CYP26B1+KCNA4SerumMSP91.392.1(113)
SLC19A3PlasmaMSRED-qPCR  85 (17/20)  85 (17/20)(114)
FAM5C/MYLKSerumMSP  77.6 (45/58)  90 (27/30)(115)
ATP4BPlasmaMSP  64 (16/25)  100 (9/9)(116)
XAF1Serumrt-MSP  83.9 (141/168)  94.3 (83/88)(117)
SOX17SerumMSP  58.9 (43/73)  100 (20/20)(118)
SPG20BloodMSP  48.8 (20/41)  100 (21/21)(119)
FLNC/THBS1/UCHL1/DLEC1Serumq-MSP  FLNC: 67 (55/82), THBS1: 63.4 (52/82), UCHL1: 56.1 (46/82), DLEC1: 80.5 (66/82),  FLNC: 93.0 (80/86), THBS1: 94.2 (81/86), UCHL1: 89.5 (77/86), DLEC1: 93.0 (80/86)(120)
OSR2/VAV3/PPFIA3SerumMSP  OSR2: 62.5 (30/48), VAV3: 45.8 (22/48), PPFIA3: 56.3 (27/48), Combined: 83.3 (40/48)  OSR2: 92 (23/25), VAV3: 100 (25/25), PPFIA3: 96 (24/25), Combined: 88 (22/25)(121)
TFPI2Tissueq-MSP6883(122)

[i] RASSF10, Ras-association domain family 10; RNF180, ring finger protein 180; SFRP1, secreted frizzled-related protein 1; IRX1, iroquois homeobox protein 1; CYP26B1, cytochrome P450 26B1; KCNA4, potassium voltage-gated channel subfamily A member 4; SLC19A3, solute carrier family 19 member 3; FAM5C, family with sequence similarity 5, member C; MYLK, myosin light chain kinase; ATP4B, ATPase H+/K+ transporting beta subunit; XAF1, XIAP-associated factor 1; SOX17, a member of the Sox family of transcription factors; SPG20, spastic paraplegia-20; FLNC, filamin-C; THBS1, thrombospondin 1; UCHL1, ubiquitin carboxy-terminal hydrolase L1; DLEC1, deleted in lung and esophageal cancer1; OSR2, protein odd-skipped-related 2; VAV3, guanine nucleotide exchange factor; PPFIA3, PTPRF-interacting protein alpha-3; TFPI2, tissue factor pathway inhibitor 2.

Current challenges

Although the genetic landscape for GC has been well researched and a large number of candidate biomarkers have been detected in blood samples in the past decades, none of these have yet progressed into clinical assays for GC. A number of challenges account for this delay.

First and foremost is the lack of biomarker validation studies demonstrating acceptable sensitivities and specificities for clinical use. In fact, most of the proposed biomarkers were identified or validated in retrospective studies with limited sample sizes. Quantification of plasma cfDNA alone, for example, is insufficient as a clinical biomarker due to its lack of specificity. On the other hand, detection of mutations or rearrangements of ctDNA seems more intriguing due to their biological relevance for tumor initiation and development. However, even the most commonly mutated genes, such as TP53 and KRAS, are typically aberrant in <50% of the cases in any particular cancer type. On this context, it is assumed that multigene panel analysis of ctDNA could lead to increased test sensitivity. However, the mutations of these genes are often located in different exons, and their abundance in circulation is generally elevated only in late-stage cancers. This impairs the detection of DNA-based sequence variations, as well as their utility in early cancer detection. For instance, in the recent CancerSEEK study, detecting mutations of 16 genes only achieved 40% sensitivity for early-stage tumors (38). By contrast, assaying DNA methylation (which is the epigenetic modification of CpG dinucleotides) is more robust and consistent than testing genetic alterations. There is accumulating evidence that cfDNA gene hypermethylation is more readily detectable than genetic mutations in patients with GC or pre-cancerous diseases such as intestinal metaplasia and dysplasia. For example, the hypermethylation of a number of genes, including RunX3, RPRM and RASSF1A, was significantly elevated in plasma samples of patients with early-stage GC. Aside from Epi proColon, the first FDA-approved gene methylation-based GC assay, Epi proLung, which tests for plasma SHOX2 and PTGER4 gene methylation, recently received a CE-IVD mark in Europe for lung cancer detection as well (87). These developments demonstrate the current utility and future potential of cfDNA methylation assays for cancer detection.

In addition to testing sensitivity and specificity, tracking tumor location is another challenge for cfDNA-based tests. For instance, although hypermethylation of Septin 9 is preferentially detected in patients with CRC, it is also present in some patients with primary lung and stomach cancer (88,89). In fact, even CancerSEEK had to rely on conventional protein tumor biomarkers to track the tissue of origin. However, in patients with early-stage cancer, conventional protein tumor biomarkers can be difficult to detect. Recent studies using computer based-analyses of genome-wide methylation signatures have demonstrated certain potential for identifying the presence, type and location of tumors (9093). For instance, methylated haplotype load, an analysis of tissue-specific methylation haplotype blocks using whole-genome bisulfite sequencing (WGBS) data, can help to identify cancer-associated biomarkers in both tissue and plasma samples (94). Preliminary data from this analysis demonstrated its potential in determining tissue of origin as well as in predicting cancer development and progression from plasma samples of patients with lung cancer and CRC. Similarly, Kang et al (92) developed CancerLocator, a probabilistic approach to WGBS analysis that is used to predict disease burden and the tissue of origin of ctDNA based on the genome-wide methylation profile of its cfDNA. However, further prospective studies with larger sample sizes are required to validate its utility in clinical settings.

Besides biomarker validation, another challenge for developing cfDNA-based tests in GC is the lack of a standard consensus for experimental procedures, including sampling, storage conditions, cfDNA isolation and enrichment, data analysis and results interpretation (95). Currently, the technologies used for cfDNA detection include qPCR, next-generation sequencing (NGS), dPCR and ultra-sensitive amplification refractory mutation system (ARMS) PCR. Each of these methods has its advantages and drawbacks (96).

Recently, liquid biopsy testing by qPCR (Cobas and Therascreen) was FDA-approved for EGFR exon 19 deletions, EGFR L858R and EGFR T790M in patients with NSCLC. However, these kits have only been validated for allele frequencies of >1%, which is not sufficient when attempting to detect tumors at early stages (9799).

By contrast, dPCR has the highest testing sensitivity and suitability in liquid biopsies (100,101). Compared with the characteristics of NGS, dPCR is more cost-effective and has faster turnaround times. However, it has a lower throughput, and can only detect a limited number of known mutations at a time (102). NGS, on the other hand, is theoretically able to detect numerous gene mutations, amplifications and fusions in parallel with higher throughput, and has already been approved for tumor-tissue profiling in the clinic (103,104). Despite this, conventional NGS has relatively low-detection sensitivity and a high-error rate, thus limiting its usefulness for analyzing cfDNA, which occurs in low abundance in plasma samples. New targeted- and genome-wide-NGS approaches for liquid biopsy testing have been developed with improved sensitivity and error-suppression rates (105,106). However, the complicated process, quality control and cost-effectiveness of NGS still need to be improved for clinical applications.

On the other hand, an ultra-sensitive ARMS PCR assay (Udx-PCR, Super-ARMS) was developed, which can detect mutant ctDNA at an allele frequency of 0.1–0.02% in the background of 10–50 ng wild-type DNA (46,107). This is comparable to the detection sensitivity of dPCR, but with significantly improved robustness, cost-effectiveness and procedural ease.

Conclusion

Recent milestones in cfDNA analysis as a liquid biopsy for early cancer detection pave the way for its adoption in clinical practice. Among them, Epi proColon is the first population-based CRC screening product that detects gene methylation in plasma samples. Another study led by Chan et al (108) indicated that detection of Epstein-Barr virus DNA in plasma is effective for the screening of early asymptomatic nasopharyngeal carcinoma. Additionally, CancerSEEK has demonstrated how a single assay can screen multiple cancer types by combining ctDNA and protein biomarkers (38). Liquid biopsy draw the attention of independent libraries and commercial companies. In a recent research by GRAIL, which focused on applying cfDNA liquid biopsy to cancer early-stage diagnosis, the sensitivity of stage I–III was 67.3% (CI, 60.7–73.3%) in a pre-specified set of 12 cancer types (such as anus, bladder, colon/rectum, esophagus, head and neck, liver/bile-duct, lung, lymphoma, ovary, pancreas, plasma cell neoplasm and stomach), and 43.9% (CI, 39.4–48.5%) in all cancer types (54). In other studies on breast cancer, 358 cancer and 452 normal cases were included. The results indicated that for three types of breast cancer (triple negative, HER2-positive/hormone receptor-positive and HER2-negative), the sensitivity was 58, 40 and 15%, respectively. This sensitivity shows that cfDNA liquid biopsy is still far from ready-to-use for clinic diagnosis (109). Nonetheless, since non-invasive and low-cost cfDNA testing still plays an important role in consumer-grade cancer diagnosis and cancer treatment management, such as personalized medicine and cancer prognosis. In those studies, two available approaches for early cancer detection using liquid biopsy were presented: i) One assay for one cancer (one-to-one); and ii) one assay for multiple cancer types (one-to-many) by utilizing genome-wide profiling or a large genetic signature panel.

Although numerous potential biomarkers have already been identified in GC, the development of a new generation of minimally invasive cfDNA-based tests for GC early detection must consider their clinical validity and utility. These require collaborative efforts in two areas: i) Developing new assays with improved sensitivity, reproducibility, procedural standardization and cost-effectiveness; and ii) validating emerging biomarkers in larger prospective clinical studies. Although the ‘one-to-many’ liquid biopsy approach is more attractive in the long term, it presents greater challenges than the ‘one-to-one’ approach. With these recent advances in cancer genetics and assay modalities, particularly the clinical implementation of circulating methylated DNA-based CRC and lung cancer screening tests, it is expected that a new, clinically effective, liquid biopsy assay for early detection of GC will be available in the near future.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

ZH performed researched data and wrote this manuscript, HZ contributed to discussions of content and helped to draft the manuscript. BY revised the draft of the manuscript. DY designed the study and revised 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 

Global Burden of Disease Cancer Collaboration, ; Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, Dicker DJ, Chimed-Orchir O, Dandona R, et al: Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: A systematic analysis for the global burden of disease study. JAMA Oncol. 3:524–548. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Karimi P, Islami F, Anandasabapathy S, Freedman ND and Kamangar F: Gastric cancer: Descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev. 23:700–713. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Takahashi T, Saikawa Y and Kitagawa Y: Gastric cancer: Current status of diagnosis and treatment. Cancers (Basel). 5:48–63. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A, et al: Treatment of gastric cancer. World J Gastroenterol. 20:1635–1649. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Rosati G, Ferrara D and Manzione L: New perspectives in the treatment of advanced or metastatic gastric cancer. World J Gastroenterol. 15:2689–2692. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Hamashima C, Shibuya D, Yamazaki H, Inoue K, Fukao A, Saito H and Sobue T: The Japanese guidelines for gastric cancer screening. Jpn J Clin Oncol. 38:259–267. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Choi KS, Jun JK, Lee HY, Park S, Jung KW, Han MA, Choi IJ and Park EC: Performance of gastric cancer screening by endoscopy testing through the national cancer screening program of Korea. Cancer Sci. 102:1559–1564. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Leung WK, Wu MS, Kakugawa Y, Kim JJ, Yeoh KG, Goh KL, Wu KC, Wu DC, Sollano J, Kachintorn U, et al: Screening for gastric cancer in Asia: Current evidence and practice. Lancet Oncol. 9:279–287. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Ley C, Mohar A, Guarner J, Herrera-Goepfert R, Figueroa LS, Halperin D and Parsonnet J: Screening markers for chronic atrophic gastritis in Chiapas, Mexico. Cancer Epidemiol Biomarkers Prev. 10:107–112. 2001.PubMed/NCBI

10 

Katai H and Sano T: Early gastric cancer: Concepts, diagnosis, and management. Int J Clin Oncol. 10:375–383. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Tashiro A, Sano M, Kinameri K, Fujita K and Takeuchi Y: Comparing mass screening techniques for gastric cancer in Japan. World J Gastroenterol. 12:4873–4874. 2006.PubMed/NCBI

12 

Tonouchi H, Mohri Y, Kobayashi M, Tanaka K, Ohi M and Kusunoki M: Laparoscopy-assisted distal gastrectomy with laparoscopic sentinel lymph node biopsy after endoscopic mucosal resection for early gastric cancer. Surg Endosc. 21:1289–1293. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Kusano C, Iwasaki M, Kaltenbach T, Conlin A, Oda I and Gotoda T: Should elderly patients undergo additional surgery after non-curative endoscopic resection for early gastric cancer? Long-term comparative outcomes. Am J Gastroenterol. 106:1064–1069. 2011. View Article : Google Scholar : PubMed/NCBI

14 

ASGE Standards of Practice Committee, ; Ben-Menachem T, Decker GA, Early DS, Evans J, Fanelli RD, Fisher DA, Fisher L, Fukami N, Hwang JH, et al: Adverse events of upper GI endoscopy. Gastrointest Endosc. 76:707–718. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Mukoubayashi C, Yanaoka K, Ohata H, Arii K, Tamai H, Oka M and Ichinose M: Serum Pepsinogen and Gastric Cancer Screening. Intern Med. 46:261–266. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Miki K, Ichinose M, Shimizu A, Huang SC, Oka H, Furihata C, Matsushima T and Takahashi K: Serum pepsinogens as a screening test of extensive chronic gastritis. Gastroenterol Jpn. 22:133–141. 1987. View Article : Google Scholar : PubMed/NCBI

17 

Nasrollahzadeh D, Aghcheli K, Sotoudeh M, Shakeri R, Persson EC, Islami F, Kamangar F, Abnet CC, Boffetta P, Engstrand L, et al: Accuracy and cut-off values of pepsinogens I, II and gastrin 17 for diagnosis of gastric fundic atrophy: Influence of gastritis. PLoS One. 6:e269572011. View Article : Google Scholar : PubMed/NCBI

18 

Miki K and Urita Y: Using serum pepsinogens wisely in a clinical practice. J Dig Dis. 8:8–14. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Huang YK, Yu JC, Kang WM, Ma ZQ, Ye X, Tian SB and Yan C: Significance of serum pepsinogens as a biomarker for gastric cancer and atrophic gastritis screening: A systematic review and meta-analysis. PLoS One. 10:e01420802015. View Article : Google Scholar : PubMed/NCBI

20 

Zhou J, Ma X, Bi F and Liu M: Clinical significance of circulating tumor cells in gastric cancer patients. Oncotarget. 8:25713–25720. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Liu Y, Ling Y, Qi Q, Lan F, Zhu M, Zhang Y, Bao Y and Zhang C: Prognostic value of circulating tumor cells in advanced gastric cancer patients receiving chemotherapy. Mol Clin Oncol. 6:235–242. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Schwarzenbach H, Hoon DS and Pantel K: Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 11:426–437. 2011. View Article : Google Scholar : PubMed/NCBI

23 

De Mattos-Arruda L, Olmos D and Tabernero J: Prognostic and predictive roles for circulating biomarkers in gastrointestinal cancer. Future Oncol. 7:1385–1397. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Shapiro B, Chakrabarty M, Cohn EM and Leon SA: Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer. 51:2116–2120. 1983. View Article : Google Scholar : PubMed/NCBI

25 

Sai S, Ichikawa D, Tomita H, Ikoma D, Tani N, Ikoma H, Kikuchi S, Fujiwara H, Ueda Y and Otsuji E: Quantification of plasma cell-free DNA in patients with gastric cancer. Anticancer Res. 27:2747–2751. 2007.PubMed/NCBI

26 

Park JL, Kim HJ, Choi BY, Lee HC, Jang HR, Song KS, Noh SM, Kim SY, Han DS and Kim YS: Quantitative analysis of cell-free DNA in the plasma of gastric cancer patients. Oncol Lett. 3:921–926. 2012.PubMed/NCBI

27 

Kim K, Shin DG, Park MK, Baik SH, Kim TH, Kim S and Lee S: Circulating cell-free DNA as a promising biomarker in patients with gastric cancer: Diagnostic validity and significant reduction of cfDNA after surgical resection. Ann Surg Treat Res. 86:136–142. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Qian C, Ju S, Qi J, Zhao J, Shen X, Jing R, Yu J, Li L, Shi Y, Zhang L, et al: Alu-based cell-free DNA: A novel biomarker for screening of gastric cancer. Oncotarget. 8:54037–54045. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Kolesnikova EV, Tamkovich SN, Bryzgunova OE, Shelestyuk PI, Permyakova VI, Vlassov VV, Tuzikov AS, Laktionov PP and Rykova EY: Circulating DNA in the blood of gastric cancer patients. Ann N Y Acad Sci. 1137:226–231. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Coimbra S, Catarino C, Costa E, Oliveira H, Figueiredo A, Rocha-Pereira P and Santos-Silva A: Circulating cell-free DNA levels in Portuguese patients with psoriasis vulgaris according to severity and therapy. Br J Dermatol. 170:939–942. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Chen C, Shi C, Huang X, Zheng J, Zhu Z, Li Q, Qiu S, Huang Z, Zhuang Z, Wu R, et al: Molecular profiles and metastasis markers in Chinese patients with gastric carcinoma. Sci Rep. 9:139952019. View Article : Google Scholar : PubMed/NCBI

32 

Wu R, Li Q, Wu F, Shi C and Chen Q: Comprehensive analysis of CDC27 related to peritoneal metastasis by whole exome sequencing in gastric cancer. Onco Targets Ther. 13:3335–3346. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Wang R, Song S, Harada K, Ghazanfari Amlashi F, Badgwell B, Pizzi MP, Xu Y, Zhao W, Dong X, Jin J, et al: Multiplex profiling of peritoneal metastases from gastric adenocarcinoma identified novel targets and molecular subtypes that predict treatment response. Gut. 69:18–31. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Leon SA, Shapiro B, Sklaroff DM and Yaros MJ: Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 37:646–650. 1977.PubMed/NCBI

35 

Chen K, Zhao H, Yang F, Hui B, Wang T, Wang LT, Shi Y and Wang J: Dynamic changes of circulating tumour DNA in surgical lung cancer patients: Protocol for a prospective observational study. BMJ Open. 8:e0190122018. View Article : Google Scholar : PubMed/NCBI

36 

Bachet JB, Bouché O, Taieb J, Dubreuil O, Garcia ML, Meurisse A, Normand C, Gornet JM, Artru P, Louafi S, et al: RAS mutation analysis in circulating tumor DNA from patients with metastatic colorectal cancer: The AGEO RASANC prospective multicenter study. Ann Oncol. 29:1211–1219. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, et al: Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 6:224ra242014. View Article : Google Scholar : PubMed/NCBI

38 

Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, Douville C, Javed AA, Wong F, Mattox A, et al: Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 359:926–930. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, Zhang S, Lee M, Wu J, Lim KH, et al: A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut. 61:673–684. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Qian Z, Zhu G, Tang L, Wang M, Zhang L, Fu J, Huang C, Fan S, Sun Y, Lv J, et al: Whole genome gene copy number profiling of gastric cancer identifies PAK1 and KRAS gene amplification as therapy targets. Genes Chromosomes Cancer. 53:883–894. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Park KU, Lee HE, Park DJ, Jung EJ, Song J, Kim HH, Choe G, Kim WH and Lee HS: MYC quantitation in cell-free plasma DNA by real-time PCR for gastric cancer diagnosis. Clin Chem Lab Med. 47:530–536. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Park KU, Lee HE, Nam SK, Nam KH, Park DJ, Kim HH, Kim WH and Lee HS: The quantification of HER2 and MYC gene fragments in cell-free plasma as putative biomarkers for gastric cancer diagnosis. Clin Chem Lab Med. 52:1033–1040. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Kinugasa H, Nouso K, Tanaka T, Miyahara K, Morimoto Y, Dohi C, Matsubara T, Okada H and Yamamoto K: Droplet digital PCR measurement of HER2 in patients with gastric cancer. Br J Cancer. 112:1652–1655. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Shoda K, Ichikawa D, Fujita Y, Masuda K, Hiramoto H, Hamada J, Arita T, Konishi H, Komatsu S, Shiozaki A, et al: Monitoring the HER2 copy number status in circulating tumor DNA by droplet digital PCR in patients with gastric cancer. Gastric Cancer. 20:126–135. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Taby R and Issa JP: Cancer epigenetics. CA Cancer J Clin. 60:376–392. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Li Y, Xu H, Su S, Ye J, Chen J, Jin X, Lin Q, Zhang D, Ye C and Chen C: Clinical validation of a highly sensitive assay to detect EGFR mutations in plasma cell-free DNA from patients with advanced lung adenocarcinoma. PLoS One. 12:e01833312017. View Article : Google Scholar : PubMed/NCBI

47 

Baylin SB, Herman JG, Graff JR, Vertino PM and Issa JP: Alterations in DNA methylation: A fundamental aspect of neoplasia. Adv Cancer Res. 72:141–196. 1998. View Article : Google Scholar : PubMed/NCBI

48 

Jones PA and Laird PW: Cancer-epigenetics comes of age. Nat Genet. 21:163–167. 1999. View Article : Google Scholar : PubMed/NCBI

49 

Ebrahimi V, Soleimanian A, Ebrahimi T, Azargun R, Yazdani P, Eyvazi S and Tarhriz V: Epigenetic modifications in gastric cancer: Focus on DNA methylation. Gene. 742:1445772020. View Article : Google Scholar : PubMed/NCBI

50 

Eyvazi S, Khamaneh AM, Tarhriz V, Bandehpour M, Hejazi MS, Sadat ATE and Sepehri B: CpG islands methylation analysis of CDH11, EphA5, and HS3ST2 genes in gastric adenocarcinoma patients. J Gastrointest Cancer. 51:579–583. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Donaldson J and Park BH: Circulating tumor DNA: Measurement and clinical utility. Annu Rev Med. 69:223–234. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Chen X, Gole J, Gore A, He Q, Lu M, Min J, Yuan Z, Yang X, Jiang Y, Zhang T, et al: Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat Commun. 11:34752020. View Article : Google Scholar : PubMed/NCBI

53 

Jiang P, Chan KCA and Lo YMD: Liver-derived cell-free nucleic acids in plasma: Biology and applications in liquid biopsies. J Hepatol. 71:409–421. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Liu MC, Oxnard GR, Klein EA, Swanton C, Seiden MV, Liu MC, Oxnard GR, Klein EA, Smith D, Richards D, et al: 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

55 

U.S. Food and Drug Administration (FDA), . Premarket Approval (PMA) for Epi ProColon. FDA; Silver Spring, MD: 2016

56 

Bennett KL, Karpenko M, Lin MT, Claus R, Arab K, Dyckhoff G, Plinkert P, Herpel E, Smiraglia D and Plass C: Frequently methylated tumor suppressor genes in head and neck squamous cell carcinoma. Cancer Res. 68:4494–4499. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Kreuziger LM, Porcher JC, Ketterling RP and Steensma DP: An MLL-SEPT9 fusion and t(11;17)(q23;q25) associated with de novo myelodysplastic syndrome. Leuk Res. 31:1145–1148. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Shirley M: Epi proColon® for colorectal cancer screening: A profile of its use in the USA. Mol Diagn Ther. 24:497–503. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Church TR, Wandell M, Lofton-Day C, Mongin SJ, Burger M, Payne SR, Castaños-Vélez E, Blumenstein BA, Rösch T, Osborn N, et al: Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 63:317–325. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Nian J, Sun X, Ming S, Yan C, Ma Y, Feng Y, Yang L, Yu M, Zhang G and Wang X: Diagnostic accuracy of methylated SEPT9 for blood-based colorectal cancer detection: A systematic review and meta-analysis. Clin Transl Gastroenterol. 8:e2162017. View Article : Google Scholar : PubMed/NCBI

61 

Cai L, Hood S, Kallam E, Overman D, Barker K, Rutledge D, Riojas J, Best C, Eisenberg M and Kam-Morgan L: Epi proColon®: Use of a non-invasive SEPT9 gene methylation blood test for colorectal cancer screening: A national laboratory experience. J Clin Epigenet. 4:72018. View Article : Google Scholar

62 

Sherr CJ: The pezcoller lecture: Cancer cell cycles revisited. Cancer Res. 60:3689–3695. 2000.PubMed/NCBI

63 

Weisenberger DJ: Characterizing DNA methylation alterations from the cancer genome atlas. J Clin Invest. 124:17–23. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Kanyama Y, Hibi K, Nakayama H, Kodera Y, Ito K, Akiyama S and Nakao A: Detection of p16 promoter hypermethylation in serum of gastric cancer patients. Cancer Sci. 94:418–420. 2003. View Article : Google Scholar : PubMed/NCBI

65 

Ichikawa D, Koike H, Ikoma H, Ikoma D, Tani N, Otsuji E, Kitamura K and Yamagishi H: Detection of aberrant methylation as a tumor marker in serum of patients with gastric cancer. Anticancer Res. 24:2477–2481. 2004.PubMed/NCBI

66 

Guo L, Huang C and Ji QJ: Aberrant promoter hypermethylation of p16, survivin, and retinoblastoma in gastric cancer. Bratisl Lek Listy. 118:164–168. 2017.PubMed/NCBI

67 

Abbaszadegan MR, Moaven O, Sima HR, Ghafarzadegan K, A'Rabi A, Forghani MN, Raziee HR, Mashhadinejad A, Jafarzadeh M, Esmaili-Shandiz E and Dadkhah E: p16 promoter hypermethylation: A useful serum marker for early detection of gastric cancer. World J Gastroenterol. 14:2055–2060. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Chen F, Liu X, Bai J, Pei D and Zheng J: The emerging role of RUNX3 in cancer metastasis (Review). Oncol Rep. 35:1227–1236. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Kim TY, Lee HJ, Hwang KS, Lee M, Kim JW, Bang YJ and Kang GH: Methylation of RUNX3 in various types of human cancers and premalignant stages of gastric carcinoma. Lab Invest. 84:479–484. 2004. View Article : Google Scholar : PubMed/NCBI

70 

Sakakura C, Hamada T, Miyagawa K, Nishio M, Miyashita A, Nagata H, Ida H, Yazumi S, Otsuji E, Chiba T, et al: Quantitative analysis of tumor-derived methylated RUNX3 sequences in the serum of gastric cancer patients. Anticancer Res. 29:2619–2625. 2009.PubMed/NCBI

71 

Lu XX, Yu JL, Ying LS, Han J, Wang S, Yu QM, Wang XB, Fang XH and Ling ZQ: Stepwise cumulation of RUNX3 methylation mediated by Helicobacter pylori infection contributes to gastric carcinoma progression. Cancer. 118:5507–5517. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Lin Z, Luo M, Chen X, He X, Qian Y, Lai S, Si J and Chen S: Combined detection of plasma ZIC1, HOXD10 and RUNX3 methylation is a promising strategy for early detection of gastric cancer and precancerous lesions. J Cancer. 8:1038–1044. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Shi DT, Han M, Gao N, Tian W and Chen W: Association of RASSF1A promoter methylation with gastric cancer risk: A meta-analysis. Tumour Biol. 35:943–948. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Wang YC, Yu ZH, Liu C, Xu LZ, Yu W, Lu J, Zhu RM, Li GL, Xia XY, Wei XW, et al: Detection of RASSF1A promoter hypermethylation in serum from gastric and colorectal adenocarcinoma patients. World J Gastroenterol. 14:3074–3080. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Balgkouranidou I, Matthaios D, Karayiannakis A, Bolanaki H, Michailidis P, Xenidis N, Amarantidis K, Chelis L, Trypsianis G, Chatzaki E, et al: Prognostic role of APC and RASSF1A promoter methylation status in cell free circulating DNA of operable gastric cancer patients. Mutat Res. 778:46–51. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Pimson C, Ekalaksananan T, Pientong C, Promthet S, Putthanachote N, Suwanrungruang K and Wiangnon S: Aberrant methylation of PCDH10 and RASSF1A genes in blood samples for non-invasive diagnosis and prognostic assessment of gastric cancer. PeerJ. 4:e21122016. View Article : Google Scholar : PubMed/NCBI

77 

Ohki R, Nemoto J, Murasawa H, Oda E, Inazawa J, Tanaka N and Taniguchi T: Reprimo, a new candidate mediator of the p53-mediated cell cycle arrest at the G2 phase. J Biol Chem. 275:22627–22630. 2000. View Article : Google Scholar : PubMed/NCBI

78 

Ooki A, Yamashita K, Yamaguchi K, Mondal A, Nishimiya H and Watanabe M: DNA damage-inducible gene, Reprimo functions as a tumor-suppressor and is suppressed by promoter methylation in gastric cancer. Mol Cancer Res. 11:1362–1374. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Bernal C, Aguayo F, Villarroel C, Vargas M, Díaz I, Ossandon FJ, Santibáñez E, Palma M, Aravena E, Barrientos C and Corvalan AH: Reprimo as a potential biomarker for early detection in gastric cancer. Clin Cancer Res. 14:6264–6269. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Liu L and Yang X: Implication of Reprimo and hMLH1 gene methylation in early diagnosis of gastric carcinoma. Int J Clin Exp Pathol. 8:14977–14982. 2015.PubMed/NCBI

81 

Lai J, Wang H, Luo Q, Huang S, Lin S, Zheng Y and Chen Q: The relationship between DNA methylation and Reprimo gene expression in gastric cancer cells. Oncotarget. 8:108610–108623. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Hu W, Zheng W, Liu Q, Chu H, Chen S, Kim JJ, Wu J and Si J: Diagnostic accuracy of DNA methylation in detection of gastric cancer: A meta-analysis. Oncotarget. 8:113142–113152. 2015. View Article : Google Scholar

83 

Chen X, Lin Z, Xue M, Si J and Chen S: Zic1 promoter hypermethylation in plasma DNA is a potential biomarker for gastric cancer and intraepithelial neoplasia. PLoS One. 10:e01339062015. View Article : Google Scholar : PubMed/NCBI

84 

Xue WJ, Feng Y, Wang F, Li P, Liu YF, Guo YB, Wang ZW and Mao QS: The value of serum RASSF10 hypermethylation as a diagnostic and prognostic tool for gastric cancer. Tumour Biol. 37:11249–11257. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Cheung KF, Lam CN, Wu K, Ng EK, Chong WW, Cheng AS, To KF, Fan D, Sung JJ and Yu J: Characterization of the gene structure, functional significance, and clinical application of RNF180, a novel gene in gastric cancer. Cancer. 118:947–959. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Liu C, Li N, Lu H, Wang Z, Chen C, Wu L, Liu J, Lu Y and Wang F: Circulating SFRP1 promoter methylation status in gastric adenocarcinoma and esophageal square cell carcinoma. Biomed Rep. 3:123–127. 2015. View Article : Google Scholar : PubMed/NCBI

87 

epigenomics: Epigenomics AG gets CE-IVD Mark for Lung Cancer Test Epi proLung(R). 2017.

88 

Powrózek T, Krawczyk P, Kucharczyk T and Milanowski J: Septin 9 promoter region methylation in free circulating DNA-potential role in noninvasive diagnosis of lung cancer: Preliminary report. Med Oncol. 31:9172014. View Article : Google Scholar : PubMed/NCBI

89 

Lee HS, Hwang SM, Kim TS, Kim DW, Park DJ, Kang SB, Kim HH and Park KU: Circulating methylated septin 9 nucleic Acid in the plasma of patients with gastrointestinal cancer in the stomach and colon. Transl Oncol. 6:290–296. 2013. View Article : Google Scholar : PubMed/NCBI

90 

Lehmann-Werman R, Neiman D, Zemmour H, Moss J, Magenheim J, Vaknin-Dembinsky A, Rubertsson S, Nellgård B, Blennow K, Zetterberg H, et al: Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc Natl Acad Sci USA. 113:E1826–E1834. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Snyder MW, Kircher M, Hill AJ, Daza RM and Shendure J: Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell. 164:57–68. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Kang S, Li Q, Chen Q, Zhou Y, Park S, Lee G, Grimes B, Krysan K, Yu M, Wang W, et al: CancerLocator: Non-invasive cancer diagnosis and tissue-of-origin prediction using methylation profiles of cell-free DNA. Genome Biol. 18:532017. View Article : Google Scholar : PubMed/NCBI

93 

Guo S, Diep D, Plongthongkum N, Fung HL and Zhang K and Zhang K: Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet. 49:635–642. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, Pacey S, Baird R and Rosenfeld N: Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat Rev Cancer. 17:223–238. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Heitzer E, Haque IS, Roberts CES and Speicher MR: Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet. 20:71–88. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Leung F, Kulasingam V, Diamandis EP, Hoon DS, Kinzler K, Pantel K and Alix-Panabières C: Circulating tumor DNA as a cancer biomarker: Fact or fiction? Clin Chem. 62:1054–1060. 2016. View Article : Google Scholar : PubMed/NCBI

97 

Bartels S, Persing S, Hasemeier B, Schipper E, Kreipe H and Lehmann U: Molecular analysis of circulating cell-free DNA from lung cancer patients in routine laboratory practice: A cross-platform comparison of three different molecular methods for mutation detection. J Mol Diagn. 19:722–732. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Wan R, Wang Z, Lee JJ, Wang S, Li Q, Tang F, Wang J, Sun Y, Bai H, Wang D, et al: Comprehensive analysis of the discordance of EGFR mutation status between tumor tissues and matched circulating tumor DNA in advanced non-small cell lung cancer. J Thorac Oncol. 12:1376–1387. 2017. View Article : Google Scholar : PubMed/NCBI

99 

Guttery DS, Page K, Hills A, Woodley L, Marchese SD, Rghebi B, Hastings RK, Luo J, Pringle JH, Stebbing J, et al: Noninvasive detection of activating estrogen receptor 1 (ESR1) mutations in estrogen receptor-positive metastatic breast cancer. Clin Chem. 61:974–982. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Busser B, Lupo J, Sancey L, Mouret S, Faure P, Plumas J, Chaperot L, Leccia MT, Coll JL, Hurbin A, et al: Plasma circulating tumor DNA levels for the monitoring of melanoma patients: Landscape of available technologies and clinical applications. Biomed Res Int. 2017:59861292017. View Article : Google Scholar : PubMed/NCBI

101 

Rachiglio AM, Esposito Abate R, Sacco A, Pasquale R, Fenizia F, Lambiase M, Morabito A, Montanino A, Rocco G, Romano C, et al: Limits and potential of targeted sequencing analysis of liquid biopsy in patients with lung and colon carcinoma. Oncotarget. 7:66595–66605. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Xu S, Lou F, Wu Y, Sun DQ, Zhang JB, Chen W, Ye H, Liu JH, Wei S, Zhao MY, et al: Circulating tumor DNA identified by targeted sequencing in advanced-stage non-small cell lung cancer patients. Cancer Lett. 370:324–331. 2016. View Article : Google Scholar : PubMed/NCBI

103 

Newman AM, Lovejoy AF, Klass DM, Kurtz DM, Chabon JJ, Scherer F, Stehr H, Liu CL, Bratman SV, Say C, et al: Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol. 34:547–555. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Phallen J, Sausen M, Adleff V, Leal A, Hruban C, White J, Anagnostou V, Fiksel J, Cristiano S, Papp E, et al: Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med. 9:eaan24152017. View Article : Google Scholar : PubMed/NCBI

105 

Belic J, Koch M, Ulz P, Auer M, Gerhalter T, Mohan S, Fischereder K, Petru E, Bauernhofer T, Geigl JB, et al: Rapid identification of plasma DNA samples with increased ctDNA levels by a modified FAST-SeqS approach. Clin Chem. 61:838–849. 2015. View Article : Google Scholar : PubMed/NCBI

106 

Kinde I, Wu J, Papadopoulos N, Kinzler KW and Vogelstein B: Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA. 108:9530–9535. 2011. View Article : Google Scholar : PubMed/NCBI

107 

Zhai J, Wu Y, Luo X, Li X and Yu DH: Abstract 643: An ultra-sensitive multiplex allele-specific real-time PCR (Udx-PCR) assay for detection of KRAS BRAF NRAS mutations in colorectal cancer. Cancer Res. 78:6432018.

108 

Chan KCA, Woo JKS, King A, Zee BCY, Lam WKJ, Chan SL, Chu SWI, Mak C, Tse IOL, Leung SYM, et al: Analysis of plasma epstein-barr virus DNA to screen for nasopharyngeal cancer. N Engl J Med. 377:513–522. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Fiala C and Diamandis EP: Can Grail find the trail to early cancer detection? Clin Chem Lab Med. 57:403–406. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Liu YH, Zhang LH, Ren H, Zhang GG, Qin F, Kong GZ, Deng GR and Ji JF: Promoter hypermethylation of p16 gene in pre- and post-operative plasma of patients with gastric adenocarcinoma. Beijing Da Xue Xue Bao Yi Xue Ban. 37:257–260. 2005.(In Chinese). PubMed/NCBI

111 

Koike H, Ichikawa D, Ikoma H, Otsuji E, Kitamura K and Yamagishi H: Comparison of methylation-specific polymerase chain reaction (MSP) with reverse transcriptase-polymerase chain reaction (RT-PCR) in peripheral blood of gastric cancer patients. J Surg Oncol. 87:182–186. 2004. View Article : Google Scholar : PubMed/NCBI

112 

Guo X, Liu W, Pan Y, Ni P, Ji J, Guo L, Zhang J, Wu J, Jiang J, Chen X, et al: Homeobox gene IRX1 is a tumor suppressor gene in gastric carcinoma. Oncogene. 29:3908–3920. 2010. View Article : Google Scholar : PubMed/NCBI

113 

Zheng Y, Chen L, Li J, Yu B, Su L, Chen X, Yu Y, Yan M, Liu B and Zhu Z: Hypermethylated DNA as potential biomarkers for gastric cancer diagnosis. Clin Biochem. 44:1405–1411. 2011. View Article : Google Scholar : PubMed/NCBI

114 

Ng EK, Leung CP, Shin VY, Wong CL, Ma ES, Jin HC, Chu KM and Kwong A: Quantitative analysis and diagnostic significance of methylated SLC19A3 DNA in the plasma of breast and gastric cancer patients. PLoS One. 6:e222332011. View Article : Google Scholar : PubMed/NCBI

115 

Chen L, Su L, Li J, Zheng Y, Yu B, Yu Y, Yan M, Gu Q, Zhu Z and Liu B: Hypermethylated FAM5C and MYLK in serum as diagnosis and pre-warning markers for gastric cancer. Dis Markers. 32:195–202. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Raja UM, Gopal G and Rajkumar T: Intragenic DNA methylation concomitant with repression of ATP4B and ATP4A gene expression in gastric cancer is a potential serum biomarker. Asian Pac J Cancer Prev. 13:5563–5568. 2012. View Article : Google Scholar : PubMed/NCBI

117 

Ling ZQ, Lv P, Lu XX, Yu JL, Han J, Ying LS, Zhu X, Zhu WY, Fang XH, Wang S and Wu YC: Circulating methylated XAF1 DNA indicates poor prognosis for gastric cancer. PLoS One. 8:e671952013. View Article : Google Scholar : PubMed/NCBI

118 

Balgkouranidou I, Karayiannakis A, Matthaios D, Bolanaki H, Tripsianis G, Tentes AA, Lianidou E, Chatzaki E, Fiska A, Lambropoulou M, et al: Assessment of SOX17 DNA methylation in cell free DNA from patients with operable gastric cancer. Association with prognostic variables and survival. Clin Chem Lab Med. 51:1505–1510. 2013. View Article : Google Scholar : PubMed/NCBI

119 

Zhang H, Song Y, Xia P, Cheng Y, Guo Q, Diao D, Wang W, Wu X, Liu D and Dang C: Detection of aberrant hypermethylated spastic paraplegia-20 as a potential biomarker and prognostic factor in gastric cancer. Med Oncol. 31:8302014. View Article : Google Scholar : PubMed/NCBI

120 

Wang G, Zhang W, Zhou B, Jin C, Wang Z, Yang Y, Wang Z, Chen Y and Feng X: The diagnosis value of promoter methylation of UCHL1 in the serum for progression of gastric cancer. Biomed Res Int. 2015:7410302015. View Article : Google Scholar : PubMed/NCBI

121 

Li WH, Zhou ZJ, Huang TH, Guo K, Chen W, Wang Y, Zhang H, Song YC and Chang DM: Detection of OSR2, VAV3, and PPFIA3 methylation in the serum of patients with gastric cancer. Dis Markers. 2016:57805382016. View Article : Google Scholar : PubMed/NCBI

122 

Hu H, Chen X, Wang C, Jiang Y, Li J, Ying X, Yang Y, Li B, Zhou C, Zhong J, et al: The role of TFPI2 hypermethylation in the detection of gastric and colorectal cancer. Oncotarget. 8:84054–84065. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

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
Huang Z, Zhang H, Yu B and Yu D: Cell‑free DNA as a liquid biopsy for early detection of gastric cancer (Review). Oncol Lett 21: 3, 2021
APA
Huang, Z., Zhang, H., Yu, B., & Yu, D. (2021). Cell‑free DNA as a liquid biopsy for early detection of gastric cancer (Review). Oncology Letters, 21, 3. https://doi.org/10.3892/ol.2020.12264
MLA
Huang, Z., Zhang, H., Yu, B., Yu, D."Cell‑free DNA as a liquid biopsy for early detection of gastric cancer (Review)". Oncology Letters 21.1 (2021): 3.
Chicago
Huang, Z., Zhang, H., Yu, B., Yu, D."Cell‑free DNA as a liquid biopsy for early detection of gastric cancer (Review)". Oncology Letters 21, no. 1 (2021): 3. https://doi.org/10.3892/ol.2020.12264