Abstract
Liquid biopsy, a minimally invasive procedure that causes minimal pain and complication risks to patients, has been extensively studied for cancer diagnosis and treatment. Moreover, it facilitates comprehensive quantification and serial assessment of the whole-body tumor burden. Several biosources obtained through liquid biopsy have been studied as important biomarkers for establishing early diagnosis, monitoring minimal residual disease, and predicting the prognosis and response to treatment in patients with cancer. Although the clinical application of liquid biopsy in gastric cancer is not as robust as that in other cancers, biomarker studies using liquid biopsy are being actively conducted in patients with gastric cancer. Herein, we aimed to review the role of various biosources that can be obtained from patients with gastric cancer through liquid biopsies, such as blood, saliva, gastric juice, urine, stool, peritoneal lavage fluid, and ascites, by dividing them into cellular and acellular components. In addition, we reviewed previous studies on the diagnostic, prognostic, and predictive biomarkers for gastric cancer using liquid biopsy and discussed the limitations of liquid biopsy and the challenges to overcome these limitations in patients with gastric cancer.
Keywords: Gastric cancer, Liquid biopsy, Biomarker
INTRODUCTION
Gastric cancer (GC) is the fifth most common cancer and the fourth leading cause of cancer-related death worldwide [1]. Currently, endoscopic or surgical resection and surgical resection and/or systemic chemotherapy are the primary therapies used for patients with early gastric cancer (EGC) and advanced gastric cancer (AGC), respectively. Molecular targeted therapy and immunotherapy are gradually being used in some patients; however, the overall survival (OS) of patients, particularly those with AGC, remains poor [2]. Therefore, a reliable tool for early diagnosis, monitoring recurrence, and predicting prognosis or treatment responses for patients with GC is required.
Imaging techniques and histopathological examinations are widely used to detect tumors in clinical settings. However, clinically detectable tumors have a burden of 1–5×109 tumor cells and imaging techniques can detect only a certain tumor volume. Histopathological examinations yield tumor cells or tissues through biopsy or surgery; however, invasive procedures can potentially cause pain and complications in patients. More importantly, these examinations involve evaluating tumors at specific time points and spatial locations and cannot represent the overall tumor status, reflecting tumor heterogeneity and dynamic tumor progression.
Recently, liquid biopsy, a novel tool used to detect tumors, has been extensively studied for cancer diagnosis and treatment and precision medicine for patients with cancer [3]. Liquid biopsy refers to the sampling and analysis of non-solid biological samples, such as blood and other non-blood body fluids, including saliva, breast milk, gastric juice, urine, stool, cerebrospinal fluid, pleural fluid, and ascitic fluid. Compared to traditional tissue biopsy, liquid biopsy is a minimally invasive and easily obtainable procedure. Furthermore, it may comprehensively quantify the whole-body tumor burden and facilitate the longitudinal monitoring of tumor burden and molecular changes through serial assessments (Fig. 1). The development of liquid biopsy for GC is relatively slower than that for other cancers, such as lung, breast, colorectal, and prostate cancers. However, liquid biopsy is possible using not only blood, but also saliva, gastric juice, urine, stool, peritoneal lavage fluid, and ascites from patients with GC, and cellular or acellular soluble biosources can be analyzed through these various liquid biopsy samples. Substantial evidence has shown that liquid biopsy has a broad application potential in several fields, including early diagnosis, minimal residual disease (MRD) detection, as well as recurrence, prognosis, and treatment guidance prediction in patients with GC (Fig. 2) [4,5,6,7].
Fig. 1. Liquid biopsy in a patient with cancer.
Fig. 2. Biosources and clinical application of liquid biopsy in gastric cancer.
cfDNA = cell-free DNA; CTC = circulating tumor cell; ctDNA = circulating tumor DNA; PBMC = peripheral blood mononuclear cell.
Several comprehensive reviews have recently been published on liquid biopsy for GC, but most have specifically focused on blood-based biomarkers or circulating tumor DNA (ctDNA) [8,9,10]. Here, we aim to review previous studies on liquid biopsy using various biosources including blood- and non-blood-based liquid biopsy samples in patients with GC. Finally, we discuss the limitations of liquid biopsy and the challenges in overcoming them for the successful clinical application in patients with GC.
BIOSOURCES OF LIQUID BIOPSY
Liquid biopsies contain numerous cellular and acellular components that can be used as novel targets for early diagnosis, recurrence monitoring, prognostic prediction, and treatment monitoring. The detection techniques and biomarker measurements depend on the biosource of the liquid biopsy.
Cellular components
Circulating tumor cells (CTCs)
CTCs are shed into peripheral circulation from primary or metastatic tumors. However, most CTCs entering the peripheral blood are eliminated by the immune system or die owing to shear forces in the bloodstream. Only a small fraction of CTCs with stem cell-like or epithelial-mesenchymal transition (EMT) properties can survive and migrate to other sites. Additionally, the half-life of CTCs is short, approximately 1–2.5 hours [11]. Therefore, detecting and counting CTCs is highly challenging because the peripheral blood has very few CTCs.
CTC detection methods consist of enrichment, detection, and analyses. CTCs are enriched using include physical property-based techniques, such as size, density, and charge and biological techniques, including positive selection using antibodies specific to tumor antigens such as epithelial cell adhesion molecules, cytokeratin, mucin 1, and epidermal growth factor receptor 1 or 2 and negative selection using antibodies against the common leukocyte antigen cluster of differentiation (CD) 45. The CellSearch system is the most used technology and has been approved by the Food and Drug Administration for CTC detection. CTC detection techniques include traditional polymerase chain reaction (PCR) and cellular protein detection methods such as immunofluorescence, immunohistochemistry, and fluorescence-assisted in situ hybridization.
Peripheral blood mononuclear cells (PBMCs)
PBMCs are heterogeneous population of lymphocytes (T cells, B cells, and natural killer cells), monocytes, and dendritic cells and ideal liquid biopsy approach to evaluate systemic immunity in patients with cancer. These cells are isolated from whole blood using Ficoll®, a hydrophilic polysaccharide that separates blood layers, and gradient centrifugation. PBMCs are analyzed using flow cytometry to determine the expression of cell-surface and intracellular molecules and characterize distinct single-cell types or fluorescence-activated cell sorting (FACS). FACS further adds to the degree of functionality using highly specific fluorescent-conjugate labeled antibodies. Multicolor FACS provides a multiparametric analysis of various cellular subpopulations, cell-surface antigens, and transcription factors. Recently, as tumor immunity and immunotherapy have become important for patients with cancer, biomarker studies using PBMCs have been actively conducted to analyze the tumor immune microenvironment.
Acellular components
Cell-free DNA (cfDNA) and ctDNA
cfDNA is an extracellular DNA fragment released by cells. In healthy individuals, cfDNA may be of hematopoietic origin and is typically found at low levels. Although cfDNA substantially accumulates in malignant tumors, it is rapidly cleared from the liver (half-life: approximately ≤2.5 hours) [12]. Compared to short-fragment (<150 bp) cfDNAs, long-fragment (>150 bp) cfDNAs originate from necrotic tumor cells. Thus, the presence of long-fragment cfDNAs indicates the presence of tumor cells [13]. cfDNA methylation has long been investigated as a cancer biomarker [14]. Therefore, long fragments or aberrant methylation of cfDNA have emerged as front-runners in the development of cfDNA biomarkers.
ctDNA is a tumor cell-derived fragmented cfDNA with genetic and epigenetic signatures released by different tumor subclones that accounts for <1% cfDNA, and therefore a key component for developing liquid biopsy-based biomarkers in patients with cancer (half-life: <2 hours) [8,9,10,15]. A major challenge in liquid biopsy approaches is the development of methods to detect and characterize small amounts of ctDNA in large populations of cfDNA. Although serum ctDNA concentrations are higher than plasma ctDNA concentrations, serum also contains DNA released from white blood cells [16]. Therefore, plasma-based assays are more sensitive for ctDNA isolation and analysis. PCR-based techniques, including the standard quantitative PCR, digital PCR (dPCR), droplet dPCR (ddPCR), and quantitative methylation-specific PCR, are the most used method for detecting cfDNA and ctDNA. Although PCR-based techniques inexpensive and have a high sensitivity, the number of detectable mutations is limited. Thus, next-generation sequencing (NGS)-based technologies are increasingly being used for cfDNA and ctDNA detection.
Non-coding RNAs (ncRNAs)
Recently, several studies have demonstrated that RNA biomarkers have unique advantages in the field of liquid biopsy owing to their high sensitivity, tissue specificity, and low detection cost. Circulating tumor RNAs include messenger RNA (mRNA) and ncRNA fragments, such as microRNA (miRNA), long ncRNA (lncRNA), circular RNA (circRNA), and PIWI-interacting RNA (piRNA), which originate from different cells and tissues. Although ncRNAs are not translated into a protein, they act as “regulators” of several genes and proteins. ncRNAs are usually secreted into the blood or other body fluids due to cell necrosis, apoptosis, or active secretion by tumor cells [17]. MiRNAs are small, single-stranded ncRNAs comprising 21–23 nucleotides that repress gene expression by promoting the degradation of their mRNA targets or inhibiting translation. lncRNAs are transcripts consisting of >200 nucleotides and can assume several complex secondary and tertiary structures owing to their nucleotide chain length. circRNAs form covalently closed continuous loops and are more stable than linear lncRNAs. ncRNAs can be encapsulated within extracellular vesicles or exosomes to maintain their stability in the extracellular environment and protect them from nuclease activity.
Since ncRNAs and cfDNAs are nucleic acid products, they are detected similarly. Quantitative reverse transcription PCR (qRT-PCR)-, dPCR-, ddPCR-, and NGS-based RNA sequencing are the most used methods for detecting ncRNAs. qRT-PCR is the gold standard for the quantitative detection of ncRNAs with high sensitivity, reproducibility, and accuracy.
Exosomes
Exosomes are small (30–150 nm) lipid bilayer extracellular vesicles found in almost all body fluids, secreted by large multivesicular bodies, and released into the extracellular environment through fusion in response to various pathological processes [18]. They secrete various mRNAs, ncRNAs, transmembrane or encapsulated cytoplasmic proteins, and lipids from tumor or non-tumor cells into the body fluids. Owing to their lipid bilayer structure, exosomes are relatively abundant and stable in circulation, making them potential biomarkers for early diagnosis, prognosis, treatment efficacy, and drug resistance in patients with cancer.
Exosomes are difficult to isolate with high purity owing to their unique formation and transport processes. Additionally, tumor cell-extracted exosomes account for only a small portion of the total exosomes in body fluids; thus, highly sensitive and specific detection methods are required. Exosome enrichment and detection methods are based on exosome properties such as density, size, surface composition, and precipitation. Currently, ultracentrifugation, ultrafiltration, precipitation, immunoaffinity enrichment, lipid-based separation, and microbead, microfluidic chip, or thermal enrichment are used for exosome enrichment. Western blot analysis and enzyme-linked immunosorbent assay (ELISA) are traditionally used to detect exosomes. Electron microscopy, nanoparticle tracking analysis, colorimetry, electrochemical analysis, fluorescence, clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system-assisted, and single exosome detection methods have been developed as new techniques for exosome detection.
Proteins
Proteins found in the blood have a long history of use as cancer biomarkers. For example, proteins such as cancer antigen (CA) 125 and prostate-specific antigen have been used clinically as diagnostic and prognostic biomarkers of ovarian and prostate cancer, respectively. Several serum protein biomarkers, including carcinoembryonic antigen (CEA), CA19-9, CA125, and CA72-4, have been widely used to predict prognosis, recurrence, and treatment response in patients with GC. However, they have low sensitivity and specificity, and lack GC-specific properties. Recently, several protein biomarkers, including inflammatory cytokines, chemokines, and enzymes, have been reported as diagnostic or prognostic biomarkers for patients with GC in clinical samples, such as blood, saliva, gastric juice, and stool [4,5,6,7,8,9,10]. ELISA is an economical and relatively easy method for detecting protein biomarkers.
CLINICAL SIGNIFICANCE OF LIQUID BIOPSY IN GC
Several studies on liquid biopsies of various tumor types, including GC, have shown major trend changes, as noted below. The application of liquid biopsy has shifted from early diagnostic and prognostic prediction to recurrence monitoring and treatment guidance for systemic anticancer therapy. Furthermore, the detection and analysis techniques for liquid biopsies have shifted from quantification to genomic, epigenomic, and multiomic analyses.
Blood-based liquid biopsy in GC
Blood is the most important and common source for liquid biopsies. In tumor tissue specimens, tumor heterogeneity may represent a significant problem, and blood-based specimens can be used to identify a comprehensive dynamic molecular profile for each patient with GC.
Early diagnosis
Although endoscopic surveillance is considered the standard method for GC screening in several Asian countries, this approach is inadequate for screening average-risk populations because of its invasiveness, high cost, and low compliance. Therefore, non-invasive approaches, including liquid biopsy-based molecular biomarker assays, are required. Patients with EGC or resectable AGC, who are candidates for screening for the early diagnosis, have significantly low circulating tumor-related biomarker levels in the peripheral blood. Both CTCs and ctDNAs are fragile, heterogeneous, and present in extremely small amounts. Therefore, most previous studies on early detection of circulating tumor-related diagnostic biomarkers have used highly sensitive biosources, such as cfDNA, ncRNA, or exosomal ncRNAs (Table 1) [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78].
Table 1. Blood-based diagnostic biomarkers for the early diagnosis of GC.
| Biosources | Biomarkers | No. of participants (GC/control) | Sample | Methods | Sensitivity (%) | Specificity (%) | AUC | Reference | |
|---|---|---|---|---|---|---|---|---|---|
| Cellular components | |||||||||
| CTCs | ≥2/7.5 mL | 116/31 | Whole blood | “FAST disc” centrifugal microfluidic system (size-selective isolation) | 85.3 | 90.3 | 0.928 | Kang et al. (2017) [19] | |
| EpCAM+CD44+ | 26/12 | Whole blood | Flow cytometry | 92.3 | 100 | 0.974 | Watanabe et al. (2017) [20] | ||
| Acellular components | |||||||||
| cfDNA | cfDNA level | 73/40 | Plasma | qPCR targeting the Alu repeats | NP | NP | 0.744 | Pu et al. (2016) [21] | |
| cfDNA level | 124/92 | Serum | Branched DNA-based Alu assay | 79.0 | 91.8 | 0.940 | Qian et al. (2017) [22] | ||
| cfDNA level | 428/95 | Serum | TaqMan qPCR assay | NP | NP | 0.931 | Lan et al. (2017) [23] | ||
| CancerSEEK | 1,817 (34 GC)/812 | Plasma | Multiplex PCR | 70.0 | 99.0 | 0.910 | Cohen et al. (2018) [24] | ||
| EpiPanGI Dx | 37 | Plasma | Targeted bisulfite sequencing reads | NP | NP | 0.900 | Kandimalla et al. (2021) [25] | ||
| Methylated RUNX3 | 50/61 | Serum | Combined restriction dPCR | 50.0 | 80.3 | 0.695 | Hideura et al. (2020) [26] | ||
| Methylated RPRML | 25/25 | Plasma | 5-Azacytidine assay and direct bisulfite sequencing | 56.0 | 88.0 | 0.726 | Alarcón et al. (2020) [27] | ||
| Methylated SFRP2 | 92/50 | Plasma | Bisulfite conversion kit | 60.9 | 86.0 | 0.784 | Miao et al. (2020) [28] | ||
| Methylated H19 and MALAT1 | 150/100 | Leukocytes | Bisulfite conversion kit | 69.4 (female), 65.8 (male) | 92.6 (female), 75.3 (male) | 0.821 (female), 0.741 (male) | Hu et al. (2021) [29] | ||
| miRNAs | miR-16, miR-25, miR-92a, miR-451 and miR-486-5p | 160/160 | Plasma | qRT-PCR | 72.9 | 89.2 | 0.812 | Zhu et al. (2014) [30] | |
| miR-21 | 50/50 | Serum, PBMCs | qRT-PCR | 88.4 | 79.6 | 0.912 | Wu et al. (2015) [31] | ||
| miR-627, miR-629, miR-652 | 108/96 | Plasma | qRT-PCR | 86.7 | 85.5 | 0.941 | Shin et al. (2015) [32] | ||
| miR-23b | 138/50 | Plasma | qRT-PCR | 71.0 | 74.0 | 0.800 | Zhuang et al. (2016) [33] | ||
| miR-376c | 65/108 | Plasma | qRT-PCR | 71.0 | 78.0 | 0.770 | Hung et al. (2017) [34] | ||
| miR-21, miR-93, miR-106a, miR-106b | 111/63 | Plasma | ddPCR | 84.8 (78.3*) | 79.2 (70.7%*) | 0.887 (0.809*) | Zhao et al. (2018) [35] | ||
| miR-25 | 184/56+78 | Serum | qRT-PCR | 67.3–69.4 | 80.4–81.0 | NP | Kong et al. (2019) [36] | ||
| miR-214 | 168/74 | Plasma | qRT-PCR | 73.2 | 91.9 | 0.880 | Ji et al. (2019) [37] | ||
| miR-200c | 202/250 | Serum | qRT-PCR | 74.0 | 66.0 | 0.750 | Huang et al. (2019) [38] | ||
| miR-425-5p, miR-24-3p, miR-1180-3p, miR-122-5p | 30/90 | Plasma | qRT-PCR | NP | NP | 0.829* | Zhu et al. (2019) [39] | ||
| miR-212 | 110/100 | Serum | qRT-PCR | 96.0 (94.6*) | 78.7 (60.8*) | 0.960 (0.951*) | Shao et al. (2020) [40] | ||
| hsa-miR-320a, hsa-miR-1260b, hsa-miR-6515-5p | 235/3,210 | Serum | qRT-PCR | >93.0 | >85.0 | >0.950 | Yao et al. (2021) [41] | ||
| miR-4257, miR-6785-5p, miR-187-5p, miR-5739 | 1,417/1,417 | Serum | DNA chip (3D-Gene®) | 99.6 | 95.3 | 0.998 | Abe et al. (2021) [42] | ||
| miR-18a, miR-181b, miR-335 | 176/173 | Serum | qRT-PCR | 71.6 (72.0*) | 87.9 (88.0*) | 0.860 (0.850*) | Izumi et al. (2021) [43] | ||
| miR-1306-3p, miR-936 and miR-6083, miR-659-3p, miR-6792-3p, miR-3185 | 52/30 | Plasma | qRT-PCR | NP | NP | 0.730 | Yu et al. (2021) [44] | ||
| 0.825 | |||||||||
| 12 miRNA panel | 445/4,803 | Serum | qRT-PCR | 87.0 | 68.4 | 0.848 | So et al. (2021) [45] | ||
| lncRNAs | CUDR, LSINCT-5, PTENP1 | 110/124 | Serum | qRT-PCR | 81.8 (77.8*) | 85.2 (97.0*) | 0.830 (0.830*) | Dong et al. (2015) [46] | |
| TINCR, CCAT2, AOC4P, BANCR, LINC00857 | 162/110 | Plasma | qRT-PCR | 81.0 | 86.0 | 0.900 | Zhang et al. (2017) [47] | ||
| ZNFX1-AS1 | 50/50 | Plasma | qRT-PCR | 84.0 | 68.0 | 0.850 | Xian et al. (2018) [48] | ||
| LINC00978 | 38/31 | Serum | qRT-PCR | 80.0 | 70.0 | 0.831 | Fu et al. (2018) [49] | ||
| CTC-501O10.1, AC100830.4, RP11-210K20.5 | 100/100 | Plasma | qRT-PCR | NP | NP | 0.724, 0.730, 0.737 | Liu et al. (2018) [50] | ||
| PANDAR, FOXD2-AS1, SMARCC2 | 109/106 | Plasma | qRT-PCR | NP | NP | 0.839 | Yang et al. (2019) [51] | ||
| CTC-497E21.4 | 100/84 | Serum | qRT-PCR | 81.8 | 75.0 | 0.848 | Zong et al. (2019) [52] | ||
| SNHG17 | 67/67 | Plasma | qRT-PCR | NP | NP | 0.748 | Zhang et al. (2019) [53] | ||
| ARHGAP27P1 | 53/53 | Plasma | qRT-PCR | 75.5 | 60.4 | 0.732 | Zhang et al. (2019) [54] | ||
| LINC00086 | 168/74 | Plasma | qRT-PCR | 72.6 | 83.8 | 0.860 | Ji et al. (2019) [37] | ||
| B3GALT5-AS1 | 107/87 | Serum | qRT-PCR | 64.5 | 87.4 | 0.816 | Feng et al. (2020) [55] | ||
| C5orf66-AS1 | 200/278 | Serum | qRT-PCR | 77.5 (81.2*) | 53.36 (62.9*) | 0.688 (0.789*) | Zhou et al. (2020) [56] | ||
| HCP5 | 98/82 | Serum | qRT-PCR | 80.0 | 70.0 | 0.750 | Qin et al. (2021) [57] | ||
| lnc-MB21D1-3:5, lnc-PSCA-4:2, lnc-ABCC5-2:1 | 52/30 | Plasma | qRT-PCR | NP | NP | 0.904 | Yu et al. (2021) [44] | ||
| circRNAs | hsa_circ_0000745 | 60/60 | Plasma | qRT-PCR | 85.5 | 45.0 | 0.683 | Huang et al. (2017) [58] | |
| hsa_circ_0000520 | 45/17 | Plasma | qRT-PCR | 82.4 | 84.4 | 0.900 | Sun et al. (2018) [59] | ||
| circPTPN22 | 120/104 | Plasma | qRT-PCR | 78.0 | 84.0 | 0.857 | Ma et al. (2021) [60] | ||
| 8-circRNAs | 194/94 | Serum | qRT-PCR | 89.0 (90.0*) | 62.0 (60.0*) | 0.830 (0.820*) | Roy et al. (2022) [61] | ||
| Exosomal miRNAs | miR-19b-3p, miR-106a-5p | 90/90 | Serum | mirVana PARIS Kit, TEM/western blot, qRT-PCR | 95.0 | 90.0 | 0.814 | Wang et al. (2017) [62] | |
| miR-92b-3p, let-7g-5p, miR-146b-5p, miR-9-5p | 86/62 | Serum | ExoQuick, qRT-PCR | 60.0* | 84.0* | 0.773* | Tang et al. (2020) [63] | ||
| miR-1307-3p | 70/60 | Serum | exoRNeasy Serum/Plasma Kits, NTA/TEM/western blot, qRT-PCR | NP | NP | 0.845, 0.820, 0.754, 0.732 | Ge et al. (2020) [64] | ||
| miR-590-5p | 168/50 | Serum | Ultracentrifugation, TEM/ western blot, qRT-PCR | 63.7 | 86.0 | 0.810 | Zheng et al. (2021) [65] | ||
| miR-195-5p, miR-211-5p | 88/88 | Plasma | miRNeasy Serum/Plasma Kit, NTA/TEM/western blot, qRT-PCR | NP | NP | 0.830 | Yang et al. (2021) [66] | ||
| miR-4741, miR-32, miR-3149, miR-6727 | 120/57 | Plasma | ExoQuickTM, NTA/ western blot, qRT-PCR | 0.855, 0.946, 0.768, 0.892 | Tang et al. (2022) [67] | ||||
| Exosomal lncRNAs | lncUEGC1 | 51/60 | Plasma | Ultracentrifugation, NTA/TEM, qRT-PCR | 88.2 | 88.3 | 0.876 (0.850*) | Lin et al. (2018) [68] | |
| lncRNA-HOTTIP | 126/120 | Serum | qRT-PCR | 69.8 | 85.0 | 0.827 | Zhao et al. (2018) [69] | ||
| lncRNA PCSK2-2:1 | 63/29 | Serum | HiPureExosomekits, TEM, qRT-PCR | 84.0 | 86.5 | 0.896 | Cai et al. (2019) [70] | ||
| lnc-GNAQ-6:1 | 43/27 | Serum | Particle size analysis, TEM/western blot, qRT-PCR | 83.7 | 55.6 | 0.732 | Li et al. (2020) [71] | ||
| lncRNA-GC1 | 522/219 | Serum | NanoSight particle-tracking analysis, TEM/western blot, qRT-PCR | 84.8 (88.7*) | 85.0 (80.8*) | 0.898 (0.861*) | Guo et al. (2020) [72] | ||
| Exosomal circRNAs | hsa_circ_0065149 | 39/41 | Plasma | qRT-PCR | 48.7* | 90.2* | 0.640* | Shao et al. (2020) [73] | |
| hsa_circ_0015286 | 60/30 | Plasma | ExoQuick, TEM/NTA, qRT-PCR | 82.1 | 65.7 | 0.778 | Zheng et al. (2022) [74] | ||
| Exosomal piRNAs | piR-019308, piR-004918, piR-018569 | 70/60 | Serum | exoRNeasy Serum/Plasma Kits, NTA/TEM/western blot, qRT-PCR | NP | NP | 0.820, 0.754, 0.732 | Ge et al. (2020) [64] | |
| Proteins | TFF1, TFF2, TFF3 | 183/280 | Serum | ELISA | 80.9 | 81.0 | 0.890 | Aikou et al. (2011) [75] | |
| VEGF, ADAM8, IgG to H. pylori, serum pepsinogen I, pepsinogen II | 285/238 | Serum | Luminex multiplex panel | 88.6 | 83.2 | 0.850 | Tong et al. (2016) [76] | ||
| CDH17, TFF3 | 111/44 | Plasma | ELISA | 66.7, 62.2 | 61.4, 56.8 | NP | Choi et al. (2017) [77] | ||
| TXNRD1 | 896/228 | Plasma | Ultraviolet spectrophotometry | 95.6 | 76.3 | 0.945 | Zhu et al. (2022) [78] | ||
GC = gastric cancer; AUC = area under the curve; CTC = circulating tumor cell; CD = cluster of differentiation; cfDNA = cell-free DNA; qPCR = quantitative polymerase chain reaction; dPCR = digital polymerase chain reaction; NP = not presented; miRNA = microRNA; qRT-PCR = quantitative reverse transcription polymerase chain reaction; NTA = nanoparticle tracking analysis; TEM = transmission electron microscopy; ddPCR = droplet digital polymerase chain reaction; lncRNA = long ncRNA; circRNA = circular RNA; piRNA = PIWI-interacting RNA; ELISA = enzyme-linked immunosorbent assay.
*Only early gastric cancer
Previous studies involving screening using cfDNA have traditionally measured cfDNA levels [21,22,23]; however, cfDNAs are also released from normal hematopoietic cells, not tumor-specific, and have low accuracy. Recent studies have used tumor-specific mutations or cfDNA methylation for the early diagnosis of patients with GC [24,25,26,27,28,29]. The CancerSEEK blood test uses a unique, non-invasive, multi-analyte test that simultaneously evaluates the presence of mutations and eight cancer-associated protein biomarkers in the blood [24]. CancerSEEK was used in 1,005 patients previously diagnosed with stage I–III esophageal, gastric, colorectal, liver, pancreatic, breast, lung, and ovarian cancers. Using this strategy, they identified patients with eight common cancer types with approximately 70% and 99% sensitivity and specificity, respectively. However, the accuracy of the predictions varied by tumor type and was high for colorectal, ovarian, and pancreatic cancers (up to 80%) but markedly low for gastric, liver, and lung cancers (up to 40%). EpiPanGI Dx consists of a panel of three distinct differentially methylated regions and has a diagnostic performance, with an area under the curve (AUC) of 0.850–0.950 for most gastrointestinal cancers (0.900 for GC) in cfDNA specimens [25]. Recently, several methylated cfDNAs (RUNX3, RPRML, SFRP2, H19, and MALAT1) have shown diagnostic performance in patients with GC than that in healthy participants [26,27,28,29].
ncRNAs are the most studied blood-based diagnostic biomarkers in patients with GC because of their abundance, stability, widespread circulation, and tumor-specific expression. Shin et al. have reported that the diagnostic value of the three-miRNA signature panel (miR-627, miR-629, and miR-652) was significantly higher in patients with GC than that in healthy participants, with 86.7% sensitivity, 85.5% specificity, and 0.941 AUC [32]. In a multicenter cohort of 5,248 participants from Singapore and Korea, the diagnostic value of the 12-miRNA panel had a sensitivity of 87.0%, specificity of 68.4%, and AUC of 0.848 [45]. Several studies have reported circRNA-based diagnostic biomarkers for GC, and an eight-circRNA panel was recently reported as a potential diagnostic biomarker for early GC detection [61]. Exosomal ncRNAs have recently been actively reported as diagnostic biomarkers for GC because exosomes can protect RNA from degradation by endogenous RNases, thereby enhancing exosomal ncRNA stability in peripheral blood circulation. Exosomal lncRNAs, including lncUEGC1 and lncRNA-GC1, are highly sensitive and stable biomarkers for detecting EGC [68,72].
Recurrence monitoring
Surgical resection is the primary curative treatment for locoregional GC. Although adjuvant chemotherapies, such as S-1 or capecitabine plus oxaliplatin, have improved relapse-free survival and OS, the 5-year OS rate for localized stages is approximately 60%, and approximately 40% patients with GC experience recurrence [79]. Standard methods for monitoring recurrence after surgical resection in patients with GC include radiological imaging and serum tumor marker assessment. Routine clinical tests such as endoscopy and computed tomography (CT) cannot reliably detect post-operative MRD or micrometastases after surgical resection. Commonly used serum markers, including CEA and CA19-9, detect only approximately 40% recurrences with poor sensitivity and specificity [80]. Therefore, MRD evaluation in patients with GC after surgical resection may provide an opportunity to reduce recurrence rates in resected disease if appropriate action can be taken for MRD-positive patients.
ctDNA is a reliable biomarker for detecting MRD in patients with breast, colon, and lung cancer. Recent studies have demonstrated the feasibility of using ctDNAs to detect MRD and monitor disease recurrence in patients with GC (Table 2) [83,84,85,86]. ctDNA levels can be affected by systemic and local treatments such as systemic anticancer therapy, radiotherapy, and other concurrent inflammatory processes or trauma. Therefore, blood sampling should ideally be performed at least two weeks after surgery to detect post-operative MRD. Approaches to MRD assessment are categorized into targeted tumor-informed and untargeted tumor-agnostic assessments. Tumor-informed approaches begin with genomic sequencing of the primary tumor, whereas tumor-agnostic approaches are uninformed about mutations in the primary tumor. Tumor-informed approaches are preferable because they determine MRD with high sensitivity and specificity by identifying tumor-specific variants with a remarkably low variant allele frequency (0.01%). Several studies have evaluated the role of ctDNAs in post-operative blood samples and their association with clinical outcomes in resected GC [83,84,85,86]. The presence of post-operative ctDNA significantly correlate with cancer recurrence and poor survival, and precede the radiographic detection of recurrence. Most of these studies performed targeted tumor-informed NGS and then applied them to longitudinal ctDNA monitoring after surgical resection. However, till date, few prospective large-scale studies have been conducted and consistent data are missing. Therefore, the feasibility of its routine clinical use remains challenging.
Table 2. Blood-based biomarkers for recurrence monitoring of GC.
| Biosources | Biomarkers | No. of patients with GC | Methods | Clinical significance | Reference | |
|---|---|---|---|---|---|---|
| Cellular components | ||||||
| CTCs | FR+ CTCs | 132 | CytoploRare Kit | Prediction of peritoneal metastasis | Zeng et al. (2022) [81] | |
| Acellular components | ||||||
| cfDNAs | Postoperative long-fragment LINE-1 | 99 | HELP | High risk of recurrence | Ko et al. (2021) [82] | |
| ctDNAs | Postoperative ctDNA (+) | 25 | WGS (Illumina) | High risk of recurrence | Kim et al. (2019) [83] | |
| Personalized cancer-specific rearrangements | ||||||
| Postoperative ctDNA (+) | 1,630 | 73-gene NGS (Guardant360 test) | Inferior DFS | Maron et al. (2019) [84] | ||
| Postoperative ctDNA (+) | 46 | Targeted NGS (Illumina Hiseq 3000) | High risk of recurrence | Yang et al. (2020) [85] | ||
| Postoperative ctDNA (+) | 35 (22 curative) | Targeted NGS | Poor RFS | Openshaw et al. (2020) [86] | ||
GC = gastric cancer; CTC = circulating tumor cell; cfDNA = cell-free DNA; ctDNA = circulating tumor DNA; FR = folate receptor; NGS = next-generation sequencing; HELP = HpaII tiny fragment enrichment by ligation-mediated polymerase chain reaction; WGS = whole-genome sequencing; DFS = disease-free survival; RFS = relapse-free survival.
Prognostic prediction
Prognostic biomarkers provide information on long-term outcomes irrespective of treatment, identifying patients with more aggressive tumors. It aims to predict the prognosis and adjust the treatment intensity for optimal survival rates through individual treatment for GC. Several studies have predicted the prognosis of patients with GC using blood-based biomarkers (Table 3) [33,40,69,74,82,86,88,89,90,91,92,93,94,95,96,97,98,99,100,101].
Table 3. Blood-based prognostic biomarkers of GC.
| Biosources | Biomarkers | No. of patients with GC | Methods | Clinical significance | Reference | |
|---|---|---|---|---|---|---|
| Cellular components | ||||||
| CTCs | >5 CTCs/7.5 mL | 65 | OBP-401 | Poor OS | Ito et al. (2016) [88] | |
| ≥2 CTCs/7.5 mL | 106 | CellSearch | Poor PFS and OS | Pernot et al. (2017) [89] | ||
| >3 CTCs/7.5 mL | 59 | CellSearch, CTC-Biopsy system | Poor PFS and OS | Ning et al. (2021) [90] | ||
| CD44+ CTCs | 228 | Immunofluorescent double staining | Poor OS | Szczepanik et al. (2019) [91] | ||
| CSV+PD-1+ CTCs | 70 | Microbead selection method | Poor OS | Liu et al. (2020) [92] | ||
| TWIST+ CTCs | 31 | Centrifugal microfluidic system | Poor OS | Jhi et al. (2021) [93] | ||
| PBMCs | High CD3+CD8+ T-cells, low CD4+CD25+Foxp3+ Tregs | 105 | FACS | Increased OS | He et al. (2017) [94] | |
| TIM-3+CD8+ T-cells | 47 | FACS | Increased OS | Ma et al. (2023) [95] | ||
| Early increase in PD-1+CD8+ T-cells after chemotherapy | 68 | FACS | Increased OS | Shin et al. (2023) [96] | ||
| Acellular components | ||||||
| cfDNAs | cfDNA level | 277 | TaqMan qPCR | Peritoneal recurrence and poor OS | Fang et al. (2016) [97] | |
| Pre-surgical low methylation levels of LINE-1 | 99 | HELP | Poor RFS and OS | Ko et al. (2021) [82] | ||
| Methylation of RASSF1A, SOX17 and Wif-1 | 70 | Methylation-specific PCR | Poor PFS and OS | Karamitrousis et al. (2021) [98] | ||
| ctDNAs | ctDNA level | 277 | TaqMan qPCR (68 mutations in 8 genes) | Peritoneal recurrence and poor OS | Fang et al. (2016) [97] | |
| ctDNA level | 35 (13 palliative) | Targeted NGS | Poor OS | Openshaw et al. (2020) [86] | ||
| TP53 mutation and MET amplification | 23 | Multigene NGS-panel technology | Poor OS | Li et al. (2021) [99] | ||
| SFRP2 hypermethylation | 148 | Methylation-specific dPCR | Poor PFS and OS | Yan et al. (2021) [100] | ||
| miRNAs | High miR-23b level | 138 | qRT-PCR | Poor clinical outcome | Zhuang et al. (2016) [33] | |
| Low miR-212 level | 110 | qRT-PCR | Poor OS | Shao et al. (2020) [40] | ||
| Low miR-148a level | 132 | qRT-PCR | Poor OS | Komatsu et al. (2021) [101] | ||
| Exosomal lncRNAs | High lncRNA-HOTTIP level | 126 | qRT-PCR | Poor OS | Zhao et al. (2018) [69] | |
| Exosomal circRNAs | High hsa_circ_0015286 level | 60 | ExoQuick, TEM/NTA, qRT-PCR | Poor OS | Zheng et al. (2022) [74] | |
GC = gastric cancer; CTC = circulating tumor cell; OS = overall survival; PFS = progression-free survival; CD = cluster of differentiation; PD-1 = programmed cell death-1; PBMC = peripheral blood mononuclear cell; FACS = fluorescence-activated cell sorting; TIM-3 = T-cell immunoglobulin and mucin-domain containing-3; cfDNA = cell-free DNA; qPCR = quantitative polymerase chain reaction; HELP = HpaII tiny fragment enrichment by ligation-mediated polymerase chain reaction; RFS = relapse-free survival; NGS = next-generation sequencing; dPCR = digital polymerase chain reaction; miRNA = microRNA; qRT-PCR = quantitative reverse transcription polymerase chain reaction; lncRNA = long non-coding RNA; circRNA = circular RNA; TEM = transmission electron microscopy; NTA = nanoparticle tracking analysis.
CTCs are traditional prognostic biomarkers of liquid biopsy and have been reported for the longest time to predict prognosis in several cancer types. CTC count significantly correlates with poor OS in patients with GC. In a meta-analysis of 579 patients with GC from 7 studies, CTC detection in peripheral blood is significantly associated with poor OS and progression-free survival (PFS) [87]. However, the detection rate and cut-off value of CTCs differed in each study [88,89,90]. CTCs that express specific molecules are potential prognostic biomarkers in patients with GC [91,92,93]. CTCs expressing stem cell markers (CD44) [91] or mesenchymal markers (cell-surface vimentin or TWIST) [92,93] are significantly associated with poor OS in patients with GC.
cfDNAs or ctDNAs can reflect the tumor status in almost real time because they have a short half-life and can serve as prognostic biomarkers for patients with GC. High cfDNA or ctDNA levels are associated with a poor prognosis [86,97]. The methylation status of several genes in the cfDNA (LINE-1, RASSF1A, SOX17, and Wif-1) and ctDNA (SFRP2) is significantly associated with PFS and OS [82,98,100]. Only few studies on miRNAs or exosomal ncRNAs have investigated the correlation between ncRNAs and survival of patients with GC. High miR-23b, exosomal lncRNA-HOTTIP, and exosomal hsa_circ_0015286 levels [33,69,74] and low miR-212 and miR-148a levels [40,101] are associated with poor OS in patients with GC.
Since the tumor immune microenvironment is closely related to the prognosis of patients with cancer, studies have focused on predicting the prognosis of patients with cancer by analyzing immune cells and tumor-related components [94,95,96]. Patients with high CD3+CD8+ T cells and low CD4+CD25+Foxp3+ regulatory T cells after neoadjuvant chemotherapy in PBMCs had significantly increased OS in patients with GC receiving neoadjuvant chemotherapy [94]. Ma et al. [95] reported that the percentage of T-cell immunoglobulin and mucin-domain containing-3 (TIM-3)+CD8+ T cells is an independent protective factor in patients with GC. Shin et al. [96] reported that an early increase in programmed cell death-1 (PD-1)+CD8+ T cells potentially predicts favorable prognosis and durable responses in patients with GC receiving palliative platinum-based chemotherapy. Therefore, high expression of immune checkpoint receptors such as PD-1 or TIM-3 in tumor-infiltrating lymphocytes reflects T-cell exhaustion, whereas an elevated proportion of T cells expressing immune checkpoint receptors in PBMCs may reflect systemic immune activation.
Treatment guidance
The area showing the greatest change in standard treatment for GC is the development of novel anticancer drugs. In addition to conventional cytotoxic chemotherapy, molecular targeted therapy and immunotherapy have significantly improved the survival of patients with advanced or metastatic GC [2]. However, the limitations of systemic anticancer therapy include primary resistance, which does not respond from the start of treatment, and secondary resistance, which acquires resistance while maintaining tumor response. Additionally, the overall tumor status may change over time after systemic anticancer therapy is administered. Therefore, dynamic monitoring through sequential liquid biopsies is essential for predicting drug response or resistance, especially in patients with GC receiving palliative anticancer systemic therapy (Table 4) [102,103,104,105,106,107,108,109,110,111,112,113,114].
Table 4. Blood-based predictive biomarkers for the treatment guidance of GC.
| Biosources | Biomarkers | No. of patients with GC | Methods | Clinical significance | Reference | |
|---|---|---|---|---|---|---|
| Cellular components | ||||||
| CTCs | ≥4 CTCs at 2 and 4 weeks | 52 | CellSearch system (Veridex) | Determination of tumor response to S-1-based or paclitaxel regimens | Matsusaka et al. (2010) [102] | |
| ≥2 HER2+ CTCs/6 mL | 115 (56 HER2+) | Metafer-iFISH Cytelligen system | Resistance to trastuzumab | Li et al. (2018) [103] | ||
| CTC-PD-L1+ | 32 | CanPatrol® CTC enrichment technique | Guidance of immunotherapy | Cheng et al. (2019) [104] | ||
| EMT progression of single CTC | 24 | Size selective method | Detection of metastasis and drug resistance for systemic therapy | Negish et al. (2022) [105] | ||
| Single-cell RNA sequencing | ||||||
| PBMCs | CD103+PD-1+CD8+ T cells | 29 | FACS | Prediction of anti-PD-1 therapy | Nose et al. (2023) [106] | |
| Acellular components | ||||||
| cfDNA | cfDNA MSI-H | 16 | Pan-cancer MSI detection using Guardant360 (74-gene panel) | Concordant with tissue MSI | Willis et al. (2019) [107] | |
| Response prediction of immunotherapy | ||||||
| cfDNA level | 106 | SuperbDNA™ hybridization | Monitoring of the efficacy of systemic therapy | Zhong et al. (2020) [108] | ||
| ctDNA | ctDNA level 3 months after the start of chemotherapy | 30 | GeneQuant RNA/DNA Calculator-Amersham Pharmacia Biotech (Biochrom) Ltd. | Correlated with DFS for chemotherapy | Normando et al. (2018) [109] | |
| ctDNA levels 6 weeks post-treatment | 61 | NGS (73-gene sequencing panel, Guardant360) | Prediction of response to anti-PD-1 and PFS | Kim et al. (2018) [110] | ||
| Genomic alterations | 55 | Targeted NGS (54–73 gene panel) | Targetable alterations by an FDA-approved agent | Kato et al. (2018) [111] | ||
| Chromosomal instability assessed by CNI score of ctDNA | 55 | WGS | Prediction of the tumor response of systemic therapy | Chen et al. (2019) [112] | ||
| Higher mTBI (≥1%) in pretreatment ctDNA | 39 HER2+ | NGS | Poor PFS | Wang et al. (2019) [113] | ||
| HER2 SCNA, PIK3CA, ERBB2/4, NF1 mutations | 24 HER2+ | Targeted NGS | Prediction of trastuzumab resistance | Wang et al. (2019) [114] | ||
GC = gastric cancer; CTC = circulating tumor cell; HER2 = human epidermal growth factor receptor 2; PD-L1 = programmed death-ligand 1; EMT = epithelial-mesenchymal transition; PBMC = peripheral blood mononuclear cell; PD-1 = programmed cell death-1; FACS = fluorescence-activated cell sorting; cfDNA = cell-free DNA; MSI = microsatellite instability; ctDNA = circulating tumor DNA; DFS = disease-free survival; NGS = next-generation sequencing; FDA = Food and Drug Administration; CNI = copy number instability; WGS = whole-genome sequencing; mTBI = molecular tumor burden index; PFS = progression-free survival.
The number of CTCs, EMT progression of single CTCs, ctDNA levels, and chromosomal instability of ctDNA are related to tumor response or survival in patients with GC receiving systemic anticancer therapy [102,105,108,109,112]. Recent studies have reported predictive biomarkers that can predict the response to anti-PD-1 and trastuzumab, an anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody, therapy using blood-based liquid biopsy. The HER2 phenotype and chromosome 8 aneuploidies in CTCs, high molecular tumor burden index or HER2 somatic copy number alteration, PIK3CA, ERBB2/4, and NF1 mutations in ctDNA are associated with trastuzumab resistance [103,113,114]. Since anti-PD-1 therapies, such as nivolumab or pembrolizumab, increase survival in locally advanced unresectable or metastatic GC, the importance and need for biomarkers to predict tumor response to anti-PD-1 therapy is rapidly increasing. Kim et al. [110] have reported that changes in ctDNA levels 6 weeks after treatment predicts tumor response and the decreased ctDNA is associated with improved PFS in patients with GC treated with pembrolizumab as a salvage treatment. Programmed death-ligand 1 (PD-L1) expression and microsatellite instability (MSI) status in tumor tissues are well-known predictive biomarkers for immunotherapy. PD-L1 on CTCs or the MSI status of cfDNA can predict the tumor response to immunotherapy in GC [104,107]. Nose et al. [106] have reported that the tissue-resident marker CD103 present in peripheral blood PD-1+CD8+ T cells 2 weeks after nivolumab treatment in patients with GC is significantly associated with PFS, suggesting the importance of immune cell-related predictive biomarkers for immunotherapy.
Therefore, for patients with AGC who need to be treated with different anticancer drugs depending on the molecular status of their tumor, systemic anticancer therapy is expected to be increasingly selected based on liquid biopsy, which can reveal the entire tumor state at the time of drug administration, rather than the tumor tissue at the time of initial diagnosis.
Non-blood-based liquid biopsy in GC
Although most studies have focused on blood-based specimens as the standard concept for liquid biopsy, non-blood-based body fluids, such as saliva, gastric juice, urine, stool, and ascitic fluids, can be potential cancer biomarker sources (Tables 5 and 6) [34,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146]. However, higher heterogeneity was observed in non-blood-based body fluids than in blood-based body fluids. Therefore, the specimen collection and processing methods should be standardized to increase the comparability and clinical implementation of non-blood-based body fluids as biomarkers in patients with GC.
Table 5. Non-blood-based diagnostic biomarkers of GC from saliva, gastric juice, urine, and stool.
| Biosources | Biomarkers | No. of participants (GC/control) | Methods | Sensitivity (%) | Specificity (%) | AUC | Reference | |
|---|---|---|---|---|---|---|---|---|
| Saliva | ||||||||
| miRNAs + mRNAs | 3 mRNAs (SPINK7, PPL, and SEMA4B) and 2 miRNAs (miR140-5p and miR301a) | 163/131 | qRT-qPCR | 75.0 | 83.0 | 0.810 | Li et al. (2018) [115] | |
| mRNAs | mRNAs (SPINK7, PPL, SEMA4B, SMAD4) | 200/200 | Luminex xMAP assay + qRT-PCR | 94.0 | 91.0 | NP | Xu et al. (2020) [116] | |
| Proteins | CSTB, TPI1, and deleted in DMBT1 | 40/40 | TMT technology | 85.0 | 80.0 | 0.930 | Xiao et al. (2016) [117] | |
| Protein glycosylation | SBA and VVA | 87/134 | Lectin microarrays | 96.0 | 80.0 | 0.890 | Shu et al. (2017) [118] | |
| Microbiota | Bacterial genera | 99/194 | 16S rRNA gene sequencing | NP | NP | 0.910 | Huang et al. (2021) [119] | |
| Gastric juice | ||||||||
| miRNAs | miR-129-1-3p, miR-129-2-3p | 42/99 | qRT-PCR | 68.7 | 71.9 | 0.656 | Yu et al. (2013) [120] | |
| miR-21, miR-106a | 42/99 | qRT-PCR | 85.7, 73.8 | 97.8, 89.3 | 0.969, 0.871 | Cui et al. (2013) [121] | ||
| miR-133a | 62/142 | qRT-PCR | 85.9 | 84.8 | 0.907 | Shao et al. (2016) [122] | ||
| lncRNAs | AA174084 | 39/92 | qRT-PCR | 46.0 | 93.0 | 0.848 | Shao et al. (2014) [123] | |
| RMRP | 39/92 | qRT-PCR | 56.4 | 75.4 | 0.699 | Shao et al. (2016) [124] | ||
| ABHD11-AS1 | 39/92 | qRT-PCR | 41.0 | 93.4 | 0.650 | Yang et al. (2016) [125] | ||
| piRNAs | piR-1245 | 66/66 | qRT-PCR | 90.9 | 74.2 | 0.885 | Zhou et al. (2020) [126] | |
| Exosomal DNAs | BARHL2 methylation | 20/10 | ExoQuick-TC, Bisulfite PCR | 90.0 | 100.0 | 0.923 | Yamamoto et al. (2016) [127] | |
| Urine | ||||||||
| DNA | 8-OHdG and 8-OHG | 60/70 | SPE and UPLC-MS/MS | NP | NP | 0.777 and 0.841 | Chen et al. (2020) [128] | |
| miRNAs | miR-376c | 65/108 | qRT-PCR | 60.0 | 64.0 | 0.700 | Hung et al. (2017) [34] | |
| miR-6807-5p, miR-6856-5p, and Helicobacter pylori | 153/153 | qRT-PCR | 76.9 | 88.9 | 0.885 | Iwasaki et al. (2019) [129] | ||
| MMP-9/NGAL and ADAM12 | 35/35 | NP | NP | 0.825 | Shimura et al. (2015) [130] | |||
| Proteins | Endothelial lipase | 90/57 | Western blotting | 79.0 | 100.0 | 0.970 | Dong et al. (2013) [131] | |
| TFF1, ADAM12, H. pylori (male) | 144/138 | Quantitative proteomics analysis | NP | NP | 0.858 (male), 0.893 (female) | Shimura et al. (2020) [132] | ||
| TFF1, uBARD1, H. pylori (female) | ||||||||
| Stool | ||||||||
| Microbiome | Veillonella, Megasphaera, and Prevotella 7 genus and Streptococcus salivarius subsp. Salivarius, Bifidobacterium dentium, and Lactobacillus salivarius | 134/58 | GC-MS | NP | NP | 0.694–0.837 | Wu et al. (2020) [133] | |
| Desulfovibrio, Escherichia, Faecalibacterium, or Oscillospira | 38/35 | qPCR | NP | NP | 0.900–0.920 | Liu et al. (2021) [134] | ||
GC = gastric cancer; AUC = area under the curve; miRNA = microRNA; mRNA = messenger RNA; qRT-qPCR = quantitative reverse transcription quantitative real-time polymerase chain reaction; qRT-PCR = quantitative reverse transcription polymerase chain reaction; NP = not presented; TMT = Tandem Mass Tags; lncRNA = long non-coding RNA; piRNA = PIWI-interacting RNA; PCR = polymerase chain reaction; SPE = solid phase extraction; UPLC-MS/MS = ultra performance liquid chromatography-tandem mass spectrometry; GC-MS = gas chromatography-mass spectrometry; qPCR = quantitative polymerase chain reaction.
Table 6. Non-blood-based GC biomarkers from peritoneal lavage or ascitic fluid.
| Biosources | Biomarkers | No. of patients with GC | Methods | Clinical significance | Reference | |
|---|---|---|---|---|---|---|
| Peritoneal lavage fluid | ||||||
| CTCs | Positive CTCs | 136 | CellSearch | Poor PFS | Okabe et al. (2015) [135] | |
| Single cell suspension | Phenotypes of lymphocyte and macrophage | 122 | FACS | Peritoneal metastasis | Takahashi et al. (2022) [136] | |
| cfDNAs | Methylated THBS1 | 92 | Quantitative methylation-specific PCR | Poor DFS | Hu et al. (2021) [137] | |
| BNIP3, CHFR, CYP1B1, MINT25, SFRP2, and RASSF2 methylation | 107 | Quantitative methylation-specific PCR | Peritoneal recurrence | Hiraki et al. (2011) [138] | ||
| MINT2 methylation | 92 | Real-time methylation-specific PCR | Prediction of peritoneal metastasis | Han et al. (2014) [139] | ||
| Poor DFS | ||||||
| Exosomal miRNAs | High miR-21 and miR-1225-5p level | 24 | Ultracentrifugation, Micro BCA Protein Assay kit, qRT-PCR | Peritoneal recurrence | Tokuhisa et al. (2015) [140] | |
| Low miR-29s level | 85 | Nanosight LM10, NTA/western blot, qRT-PCR | Peritoneal recurrence | Ohzawa et al. (2020) [141] | ||
| Poor OS | ||||||
| Cytokines | IL-6 | 145 | ELISA | Peritoneal metastasis | Yang et al. (2022) [142] | |
| Ascites | ||||||
| Single cell suspension | CD45RO+CD8+ T cells, HLA-DR+CD8+ T cells, PD-1+CD3+ T cells, FoxP3+ Tregs | 55 | FACS | Poor OS | Park et al. (2019) [143] | |
| DNA, RNA | SEs at the ELF3, KLF5, and EHF loci, TGF-β pathway activation through SMAD3 SE activation and high TEAD1 expression | 98 | WGS, RNA sequencing | Molecular guided therapy by two distinct molecular subtypes of peritoneal metastases | Tanaka et al. (2021) [144] | |
| RNA | 12-gene prognostic signature | 15 | Single cell RNA sequencing | Prognostic signature of gastric dominant and GI-mixed type | Wang et al. (2021) [145] | |
| Exosomal miRNAs | Low miR-181b-5p level | 92 | miRCURY Exosome Isolation Kit, qRT-PCR | Diagnosis of GC-associated malignant ascites (AUC 0.846) | Yun et al. (2019) [146] | |
GC = gastric cancer; CTC = circulating tumor cell; PFS = progression-free survival; FACS = fluorescence-activated cell sorting; cfDNA = cell-free DNA; PCR = polymerase chain reaction; DFS = disease-free survival; miRNA = microRNA; qRT-PCR = quantitative reverse transcription polymerase chain reaction; NTA = nanoparticle tracking analysis; OS = overall survival; ELISA = enzyme-linked immunosorbent assay; WGS = whole-genome sequencing; GI = gastrointestinal; AUC = area under the curve.
Saliva
Saliva is a complex fluid containing proteins, metabolites, DNA, mRNA, ncRNAs, and microbiota that can be used as biomarkers. Using saliva as a biosource for biomarker analysis is another notable approach in patients with GC because its collection is minimally invasive and the salivary glands may be stimulated by mediators that can be released by GC. In a previous study, a salivary panel of 3 mRNAs (SPINK7, PPL, and SEMA4B) and 2 miRNAs (miR-140-5p and miR-301a) showed diagnostic performance with 75.0% sensitivity, 83.0% specificity, and 0.810 AUC in a cohort of 163 and 131 patients with and without GC, respectively [115]. Using another panel of four mRNAs (SPINK7, PPL, SEMA4B, and SMAD4), patients with GC were discriminated from healthy participants with 94.0% sensitivity and 91.0% specificity using saliva samples from 200 patients with GC and 200 healthy participants [116]. Huang et al. [119] have reported a distinct salivary microbiota as a diagnostic biomarker and demonstrated that microbiota profiles in saliva have diagnostic potential, with 0.910 AUC, in patients with GC.
However, saliva-based GC diagnosis remains unsatisfactory because of unstable quality control, oral microorganism disturbance, and salivary sample contamination with other molecules.
Gastric juice
Gastric juice is a potential specimen for GC screening and diagnosis because it exists only in the stomach and can be collected directly from the lesion area. Moreover, it can be easily obtained during diagnostic endoscopy to screen healthy participants or monitor patients for benign or precancerous lesions.
As DNA is easily degraded due to the acidity of gastric juice, most biomarkers identified in gastric juice are ncRNAs and proteins. Several miRNAs (miR-129-1-3p, miR-129-2-3p, miR-21, miR-106a, and miR-133a) have shown diagnostic performance in distinguishing GC patients from healthy participants [120,121,122]. Other ncRNAs, including lncRNAs, circRNAs, piRNAs, and exosomal RNAs, are diagnostic biomarkers for GC because of their stability in gastric juice [123,124,125,126].
Urine and stool
Urine or stool are notable biological specimens used to identify new non-invasive biomarkers for GC. Compared to blood, they can be obtained easily, in large amounts, and without invasion. Iwasaki et al. [129] found that an urinary miRNA (miR-6807-5p and miR-6856-5p) panel in combination with serum Helicobacter pylori status could distinguish patients with GC from healthy controls, with 76.9% sensitivity, 88.9% specificity, and 0.885 AUC. Recently, fecal microbiome alterations have been associated with GC and have shown good diagnostic value for distinguishing patients with GC from healthy participants [133,134].
However, the concentration of biomarkers in urine or stool has a high degree of variability and is likely to be influenced by other conditions, such as an individual’s hydration status, medications, and other comorbidities, making it difficult to determine whether these biomarkers in urine or stool are GC-specific.
Peritoneal lavage fluid and ascites
Peritoneal metastasis has an extremely poor prognosis. Therefore, accurate peritoneal metastasis diagnosis and peritoneal recurrence prediction are extremely important in patients with GC. Currently, cytological analysis of the ascitic fluid or radiological tests, such as abdominal and pelvic CT, are performed to diagnose peritoneal metastasis. However, owing to the low sensitivity of these traditional diagnostic methods, patients with GC sometimes require invasive procedures such as laparoscopy or laparotomy. Therefore, more sensitive and non-invasive diagnostic methods are required to detect peritoneal metastasis or malignant ascites in patients with GC.
Peritoneal lavage during surgical GC resection is useful for detecting free cancer cells in the peritoneal cavity. Patients with positive cytology are classified as having M1 or stage IV cancer [2]. However, cytology has low sensitivity, and several patients with GC and negative peritoneal cytology experience peritoneal recurrence after radical surgery. Previous studies have attempted to improve peritoneal recurrence prediction using RT-PCR for CEA mRNA in combination with other mRNAs. Recent studies have reported that the GC-specific cfDNA or exosomal miRNA, which are stable in the peritoneal environment, predicts peritoneal recurrence and is associated with prognosis [137,138,139,140,141,146]. Furthermore, several studies are being actively conducted to analyze malignant ascites and understand peritoneal metastasis with the development of high-quality techniques that can analyze immune cell characteristics and genomic or epigenomic changes. Park et al. [143] reported that the proportions of CD8+ T cells with memory markers (CD45RO) and activation markers (human leukocyte antigen [HLA]-DR), CD3+ T cells with PD-1, and Foxp3+ regulatory T cells profiled by FACS were significantly higher in malignant ascites than those in blood and were independent prognostic factors [143]. A comprehensive multiomic analysis of malignant ascites from 98 patients with GC revealed two distinct molecular subtypes: one displaying active super enhancers (SEs) at the ELF3, KLF5, and EHF loci, and the second displaying transforming growth factor-β pathway activation through SMAD3 SE activation and enhanced transcriptional enhancer factor-1 expression [144]. Single-cell transcriptome profiling of malignant ascites cells from 15 patients with GC divided them into two subtypes prognostically independent of clinical variables, and a 12-gene prognostic signature was derived [145].
CHALLENGES AND FUTURE PERSPECTIVES
Liquid biopsy is a powerful and innovative tool that can potentially change the diagnostic and treatment paradigm for patients with GC through early diagnosis before clinical detection, representative molecular analysis of the entire tumor, MRD detection after surgical resection, improved risk stratification, and systemic therapy guidance. However, practical clinical application of liquid biopsy in GC faces several challenges.
One of the major challenges in developing liquid biopsy for GC is its low sensitivity and specificity. Moreover, single omics information often cannot reflect the entire molecular characteristics, considering tumor heterogeneity. The integration of multiple omics information can partially complement each other and improve our understanding of tumor molecular characteristics. However, multiomics studies on liquid biopsy samples are challenging because of the remarkably low analyte content in body fluid samples. Therefore, various highly sensitive GC-specific detection methods must be developed along with liquid biopsies. In addition, specimen processing and analysis procedures should be standardized to ensure reproducibility and comparability of liquid biopsy results. Previous studies and review articles have focused on tumor-related components and blood samples. Therefore, more studies on tumor microenvironment-related components or non-blood-based samples are required. Although there are several notable biomarkers in liquid biopsies of patients with GC, the best strategy may be to combine different biomarkers and clinical and biochemical parameters to increase diagnostic or predictive performance. This approach may be one of the most feasible future strategies. Finally, clinical validation with large-scale clinical trials is necessary for its widespread use in clinical practice.
Although evidence supporting the clinical application of liquid biopsy in GC is not as robust as that in other tumor types, this field is advancing rapidly, with several active clinical trials incorporating liquid biopsy. Furthermore, liquid biopsy may be valuable for highly heterogeneous tumors, such as GC, by overcoming tumor heterogeneity and providing comprehensive molecular characterization. Taken together, liquid biopsy is an emerging tool that can play an important role in improving personalized medicine for patients with GC.
Footnotes
Funding: This review was supported by the Basic Science Research Program through the National Research Foundation of Korea, funded by the Ministry of Education, Science, and Technology (2017R1A5A2015541).
Conflict of Interest: No potential conflict of interest relevant to this article was reported.
- Conceptualization: H.H.S., L.K.W.
- Data curation: H.H.S., L.K.W.
- Formal analysis: H.H.S., L.K.W.
- Funding acquisition: H.H.S.
- Investigation: H.H.S., L.K.W.
- Methodology: H.H.S., L.K.W.
- Project administration: H.H.S., L.K.W.
- Resources: H.H.S., L.K.W.
- Software: H.H.S., L.K.W.
- Supervision: L.K.W.
- Validation: L.K.W.
- Visualization: H.H.S..
- Writing - original draft: H.H.S.
- Writing - review & editing: H.H.S., L.K.W.
References
- 1.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- 2.Kim TH, Kim IH, Kang SJ, Choi M, Kim BH, Eom BW, et al. Korean Practice Guidelines for Gastric Cancer 2022: an evidence-based, multidisciplinary approach. J Gastric Cancer. 2023;23:3–106. doi: 10.5230/jgc.2023.23.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lone SN, Nisar S, Masoodi T, Singh M, Rizwan A, Hashem S, et al. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments. Mol Cancer. 2022;21:79. doi: 10.1186/s12943-022-01543-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ma S, Zhou M, Xu Y, Gu X, Zou M, Abudushalamu G, et al. Clinical application and detection techniques of liquid biopsy in gastric cancer. Mol Cancer. 2023;22:7. doi: 10.1186/s12943-023-01715-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhang Z, Wu H, Chong W, Shang L, Jing C, Li L. Liquid biopsy in gastric cancer: predictive and prognostic biomarkers. Cell Death Dis. 2022;13:903. doi: 10.1038/s41419-022-05350-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ma X, Ou K, Liu X, Yang L. Application progress of liquid biopsy in gastric cancer. Front Oncol. 2022;12:969866. doi: 10.3389/fonc.2022.969866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chivu-Economescu M, Necula L, Matei L, Dragu D, Bleotu C, Diaconu CC. Clinical applications of liquid biopsy in gastric cancer. Front Med (Lausanne) 2021;8:749250. doi: 10.3389/fmed.2021.749250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Grizzi G, Salati M, Bonomi M, Ratti M, Holladay L, De Grandis MC, et al. Circulating tumor DNA in gastric adenocarcinoma: future clinical applications and perspectives. Int J Mol Sci. 2023;24:9421. doi: 10.3390/ijms24119421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mencel J, Slater S, Cartwright E, Starling N. The role of ctDNA in gastric cancer. Cancers (Basel) 2022;14:5105. doi: 10.3390/cancers14205105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Paschold L, Binder M. Circulating tumor DNA in gastric and gastroesophageal junction cancer. Curr Oncol. 2022;29:1430–1441. doi: 10.3390/curroncol29030120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Krebs MG, Metcalf RL, Carter L, Brady G, Blackhall FH, Dive C. Molecular analysis of circulating tumour cells-biology and biomarkers. Nat Rev Clin Oncol. 2014;11:129–144. doi: 10.1038/nrclinonc.2013.253. [DOI] [PubMed] [Google Scholar]
- 12.Thierry AR, El Messaoudi S, Gahan PB, Anker P, Stroun M. Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016;35:347–376. doi: 10.1007/s10555-016-9629-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang BG, Huang HY, Chen YC, Bristow RE, Kassauei K, Cheng CC, et al. Increased plasma DNA integrity in cancer patients. Cancer Res. 2003;63:3966–3968. [PubMed] [Google Scholar]
- 14.Luo H, Wei W, Ye Z, Zheng J, Xu RH. Liquid biopsy of methylation biomarkers in cell-free DNA. Trends Mol Med. 2021;27:482–500. doi: 10.1016/j.molmed.2020.12.011. [DOI] [PubMed] [Google Scholar]
- 15.Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–990. doi: 10.1038/nm.1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lee JS, Kim M, Seong MW, Kim HS, Lee YK, Kang HJ. Plasma vs. serum in circulating tumor DNA measurement: characterization by DNA fragment sizing and digital droplet polymerase chain reaction. Clin Chem Lab Med. 2020;58:527–532. doi: 10.1515/cclm-2019-0896. [DOI] [PubMed] [Google Scholar]
- 17.Cech TR, Steitz JA. The noncoding RNA revolution-trashing old rules to forge new ones. Cell. 2014;157:77–94. doi: 10.1016/j.cell.2014.03.008. [DOI] [PubMed] [Google Scholar]
- 18.Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32:466–477. doi: 10.1016/j.annonc.2021.01.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kang HM, Kim GH, Jeon HK, Kim DH, Jeon TY, Park DY, et al. Circulating tumor cells detected by lab-on-a-disc: role in early diagnosis of gastric cancer. PLoS One. 2017;12:e0180251. doi: 10.1371/journal.pone.0180251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Watanabe T, Okumura T, Hirano K, Yamaguchi T, Sekine S, Nagata T, et al. Circulating tumor cells expressing cancer stem cell marker CD44 as a diagnostic biomarker in patients with gastric cancer. Oncol Lett. 2017;13:281–288. doi: 10.3892/ol.2016.5432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pu WY, Zhang R, Xiao L, Wu YY, Gong W, Lv XD, et al. Prediction of cancer progression in a group of 73 gastric cancer patients by circulating cell-free DNA. BMC Cancer. 2016;16:943. doi: 10.1186/s12885-016-2977-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Qian C, Ju S, Qi J, Zhao J, Shen X, Jing R, et al. Alu-based cell-free DNA: a novel biomarker for screening of gastric cancer. Oncotarget. 2016;8:54037–54045. doi: 10.18632/oncotarget.11079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lan YT, Chen MH, Fang WL, Hsieh CC, Lin CH, Jhang FY, et al. Clinical relevance of cell-free DNA in gastrointestinal tract malignancy. Oncotarget. 2017;8:3009–3017. doi: 10.18632/oncotarget.13821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–930. doi: 10.1126/science.aar3247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kandimalla R, Xu J, Link A, Matsuyama T, Yamamura K, Parker MI, et al. EpiPanGI Dx: a cell-free DNA methylation fingerprint for the early detection of gastrointestinal cancers. Clin Cancer Res. 2021;27:6135–6144. doi: 10.1158/1078-0432.CCR-21-1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hideura E, Suehiro Y, Nishikawa J, Shuto T, Fujimura H, Ito S, et al. Blood free-circulating DNA testing of methylated RUNX3 is useful for diagnosing early gastric cancer. Cancers (Basel) 2020;12:789. doi: 10.3390/cancers12040789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Alarcón MA, Olivares W, Córdova-Delgado M, Muñoz-Medel M, de Mayo T, Carrasco-Aviño G, et al. The reprimo-like gene is an epigenetic-mediated tumor suppressor and a candidate biomarker for the non-invasive detection of gastric cancer. Int J Mol Sci. 2020;21:9472. doi: 10.3390/ijms21249472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Miao J, Liu Y, Zhao G, Liu X, Ma Y, Li H, et al. Feasibility of plasma-methylated SFRP2 for early detection of gastric cancer. Cancer Contr. 2020;27:1073274820922559. doi: 10.1177/1073274820922559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hu D, Lou X, Meng N, Li Z, Teng Y, Zou Y, et al. Peripheral blood-based DNA methylation of long non-coding RNA H19 and metastasis-associated lung adenocarcinoma transcript 1 promoters are potential non-invasive biomarkers for gastric cancer detection. Cancer Contr. 2021;28:10732748211043667. doi: 10.1177/10732748211043667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhu C, Ren C, Han J, Ding Y, Du J, Dai N, et al. A five-microRNA panel in plasma was identified as potential biomarker for early detection of gastric cancer. Br J Cancer. 2014;110:2291–2299. doi: 10.1038/bjc.2014.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wu J, Li G, Wang Z, Yao Y, Chen R, Pu X, et al. Circulating microRNA-21 is a potential diagnostic biomarker in gastric cancer. Dis Markers. 2015;2015:435656. doi: 10.1155/2015/435656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Shin VY, Ng EK, Chan VW, Kwong A, Chu KM. A three-miRNA signature as promising non-invasive diagnostic marker for gastric cancer. Mol Cancer. 2015;14:202. doi: 10.1186/s12943-015-0473-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhuang K, Han K, Tang H, Yin X, Zhang J, Zhang X, et al. Up-Regulation of Plasma miR-23b is Associated with Poor Prognosis of Gastric Cancer. Med Sci Monit. 2016;22:356–361. doi: 10.12659/MSM.895428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hung PS, Chen CY, Chen WT, Kuo CY, Fang WL, Huang KH, et al. miR-376c promotes carcinogenesis and serves as a plasma marker for gastric carcinoma. PLoS One. 2017;12:e0177346. doi: 10.1371/journal.pone.0177346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhao G, Jiang T, Liu Y, Huai G, Lan C, Li G, et al. Droplet digital PCR-based circulating microRNA detection serve as a promising diagnostic method for gastric cancer. BMC Cancer. 2018;18:676. doi: 10.1186/s12885-018-4601-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kong Y, Ning L, Qiu F, Yu Q, Cao B. Clinical significance of serum miR-25 as a diagnostic and prognostic biomarker in human gastric cancer. Cancer Biomark. 2019;24:477–483. doi: 10.3233/CBM-182213. [DOI] [PubMed] [Google Scholar]
- 37.Ji B, Huang Y, Gu T, Zhang L, Li G, Zhang C. Potential diagnostic and prognostic value of plasma long noncoding RNA LINC00086 and miR-214 expression in gastric cancer. Cancer Biomark. 2019;24:249–255. doi: 10.3233/CBM-181486. [DOI] [PubMed] [Google Scholar]
- 38.Huang ZS, Guo XW, Zhang G, Liang LX, Nong B. The diagnostic and prognostic value of miR-200c in gastric cancer: a meta-analysis. Dis Markers. 2019;2019:8949618. doi: 10.1155/2019/8949618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhu XL, Ren LF, Wang HP, Bai ZT, Zhang L, Meng WB, et al. Plasma microRNAs as potential new biomarkers for early detection of early gastric cancer. World J Gastroenterol. 2019;25:1580–1591. doi: 10.3748/wjg.v25.i13.1580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Shao JP, Su F, Zhang SP, Chen HK, Li ZJ, Xing GQ, et al. miR-212 as potential biomarker suppresses the proliferation of gastric cancer via targeting SOX4. J Clin Lab Anal. 2020;34:e23511. doi: 10.1002/jcla.23511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yao Y, Ding Y, Bai Y, Zhou Q, Lee H, Li X, et al. Identification of serum circulating microRNAs as novel diagnostic biomarkers of gastric cancer. Front Genet. 2021;11:591515. doi: 10.3389/fgene.2020.591515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Abe S, Matsuzaki J, Sudo K, Oda I, Katai H, Kato K, et al. A novel combination of serum microRNAs for the detection of early gastric cancer. Gastric Cancer. 2021;24:835–843. doi: 10.1007/s10120-021-01161-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Izumi D, Zhu Z, Chen Y, Toden S, Huo X, Kanda M, et al. Assessment of the diagnostic efficiency of a liquid biopsy assay for early detection of gastric cancer. JAMA Netw Open. 2021;4:e2121129. doi: 10.1001/jamanetworkopen.2021.21129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yu Z, Rong Z, Sheng J, Luo Z, Zhang J, Li T, et al. Aberrant non-coding RNA expressed in gastric cancer and its diagnostic value. Front Oncol. 2021;11:606764. doi: 10.3389/fonc.2021.606764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.So JB, Kapoor R, Zhu F, Koh C, Zhou L, Zou R, et al. Development and validation of a serum microRNA biomarker panel for detecting gastric cancer in a high-risk population. Gut. 2021;70:829–837. doi: 10.1136/gutjnl-2020-322065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dong L, Qi P, Xu MD, Ni SJ, Huang D, Xu QH, et al. Circulating CUDR, LSINCT-5 and PTENP1 long noncoding RNAs in sera distinguish patients with gastric cancer from healthy controls. Int J Cancer. 2015;137:1128–1135. doi: 10.1002/ijc.29484. [DOI] [PubMed] [Google Scholar]
- 47.Zhang K, Shi H, Xi H, Wu X, Cui J, Gao Y, et al. Genome-wide lncRNA microarray profiling identifies novel circulating lncRNAs for detection of gastric cancer. Theranostics. 2017;7:213–227. doi: 10.7150/thno.16044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xian HP, Zhuo ZL, Sun YJ, Liang B, Zhao XT. Circulating long non-coding RNAs HULC and ZNFX1-AS1 are potential biomarkers in patients with gastric cancer. Oncol Lett. 2018;16:4689–4698. doi: 10.3892/ol.2018.9199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fu M, Huang Z, Zang X, Pan L, Liang W, Chen J, et al. Long noncoding RNA LINC00978 promotes cancer growth and acts as a diagnostic biomarker in gastric cancer. Cell Prolif. 2018;51:e12425. doi: 10.1111/cpr.12425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Liu J, Wang J, Song Y, Ma B, Luo J, Ni Z, et al. A panel consisting of three novel circulating lncRNAs, is it a predictive tool for gastric cancer? J Cell Mol Med. 2018;22:3605–3613. doi: 10.1111/jcmm.13640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Yang Z, Sun Y, Liu R, Shi Y, Ding S. Plasma long noncoding RNAs PANDAR, FOXD2-AS1, and SMARCC2 as potential novel diagnostic biomarkers for gastric cancer. Cancer Manag Res. 2019;11:6175–6184. doi: 10.2147/CMAR.S201935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zong W, Feng W, Jiang Y, Ju S, Cui M, Jing R. Evaluating the diagnostic and prognostic value of serum long non-coding RNA CTC-497E21.4 in gastric cancer. Clin Chem Lab Med. 2019;57:1063–1072. doi: 10.1515/cclm-2018-0929. [DOI] [PubMed] [Google Scholar]
- 53.Zhang G, Xu Y, Wang S, Gong Z, Zou C, Zhang H, et al. LncRNA SNHG17 promotes gastric cancer progression by epigenetically silencing of p15 and p57. J Cell Physiol. 2019;234:5163–5174. doi: 10.1002/jcp.27320. [DOI] [PubMed] [Google Scholar]
- 54.Zhang G, Xu Y, Zou C, Tang Y, Lu J, Gong Z, et al. Long noncoding RNA ARHGAP27P1 inhibits gastric cancer cell proliferation and cell cycle progression through epigenetically regulating p15 and p16. Aging (Albany NY) 2019;11:9090–9110. doi: 10.18632/aging.102377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Feng W, Zong W, Li Y, Shen X, Cui X, Ju S. Abnormally expressed long noncoding RNA B3GALT5-AS1 may serve as a biomarker for the diagnostic and prognostic of gastric cancer. J Cell Biochem. 2020;121:557–565. doi: 10.1002/jcb.29296. [DOI] [PubMed] [Google Scholar]
- 56.Zhou Q, Li H, Jing J, Yuan Y, Sun L. Evaluation of C5orf66-AS1 as a potential biomarker for predicting early gastric cancer and its role in gastric carcinogenesis. Onco Targets Ther. 2020;13:2795–2805. doi: 10.2147/OTT.S239965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Qin S, Yang L, Kong S, Xu Y, Liang B, Ju S. LncRNA HCP5: a potential biomarker for diagnosing gastric cancer. Front Oncol. 2021;11:684531. doi: 10.3389/fonc.2021.684531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Huang M, He YR, Liang LC, Huang Q, Zhu ZQ. Circular RNA hsa_circ_0000745 may serve as a diagnostic marker for gastric cancer. World J Gastroenterol. 2017;23:6330–6338. doi: 10.3748/wjg.v23.i34.6330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sun H, Tang W, Rong D, Jin H, Fu K, Zhang W, et al. Hsa_circ_0000520, a potential new circular RNA biomarker, is involved in gastric carcinoma. Cancer Biomark. 2018;21:299–306. doi: 10.3233/CBM-170379. [DOI] [PubMed] [Google Scholar]
- 60.Ma S, Kong S, Gu X, Xu Y, Tao M, Shen L, et al. As a biomarker for gastric cancer, circPTPN22 regulates the progression of gastric cancer through the EMT pathway. Cancer Cell Int. 2021;21:44. doi: 10.1186/s12935-020-01701-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Roy S, Kanda M, Nomura S, Zhu Z, Toiyama Y, Taketomi A, et al. Diagnostic efficacy of circular RNAs as noninvasive, liquid biopsy biomarkers for early detection of gastric cancer. Mol Cancer. 2022;21:42. doi: 10.1186/s12943-022-01527-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Wang N, Wang L, Yang Y, Gong L, Xiao B, Liu X. A serum exosomal microRNA panel as a potential biomarker test for gastric cancer. Biochem Biophys Res Commun. 2017;493:1322–1328. doi: 10.1016/j.bbrc.2017.10.003. [DOI] [PubMed] [Google Scholar]
- 63.Tang S, Cheng J, Yao Y, Lou C, Wang L, Huang X, et al. Combination of four serum exosomal MiRNAs as novel diagnostic biomarkers for early-stage gastric cancer. Front Genet. 2020;11:237. doi: 10.3389/fgene.2020.00237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ge L, Zhang N, Li D, Wu Y, Wang H, Wang J. Circulating exosomal small RNAs are promising non-invasive diagnostic biomarkers for gastric cancer. J Cell Mol Med. 2020;24:14502–14513. doi: 10.1111/jcmm.16077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Zheng GD, Xu ZY, Hu C, Lv H, Xie HX, Huang T, et al. Exosomal miR-590-5p in serum as a biomarker for the diagnosis and prognosis of gastric cancer. Front Mol Biosci. 2021;8:636566. doi: 10.3389/fmolb.2021.636566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yang J, Li X, Wei S, Peng L, Sang H, Jin D, et al. Evaluation of the diagnostic potential of a plasma exosomal miRNAs panel for gastric cancer. Front Oncol. 2021;11:683465. doi: 10.3389/fonc.2021.683465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Tang G, Wang J, Dong W, Dai K, Du J. Exosomal miRNA expression profiling and the roles of exosomal miR-4741, miR-32, miR-3149, and miR-6727 on gastric cancer progression. BioMed Res Int. 2022;2022:1263812. doi: 10.1155/2022/1263812. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 68.Lin LY, Yang L, Zeng Q, Wang L, Chen ML, Zhao ZH, et al. Tumor-originated exosomal lncUEGC1 as a circulating biomarker for early-stage gastric cancer. Mol Cancer. 2018;17:84. doi: 10.1186/s12943-018-0834-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhao R, Zhang Y, Zhang X, Yang Y, Zheng X, Li X, et al. Exosomal long noncoding RNA HOTTIP as potential novel diagnostic and prognostic biomarker test for gastric cancer. Mol Cancer. 2018;17:68. doi: 10.1186/s12943-018-0817-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Cai C, Zhang H, Zhu Y, Zheng P, Xu Y, Sun J, et al. Serum exosomal long noncoding RNA pcsk2-2:1 as a potential novel diagnostic biomarker for gastric cancer. Onco Targets Ther. 2019;12:10035–10041. doi: 10.2147/OTT.S229033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Li S, Zhang M, Zhang H, Hu K, Cai C, Wang J, et al. Exosomal long noncoding RNA lnc-GNAQ-6:1 may serve as a diagnostic marker for gastric cancer. Clin Chim Acta. 2020;501:252–257. doi: 10.1016/j.cca.2019.10.047. [DOI] [PubMed] [Google Scholar]
- 72.Guo X, Lv X, Ru Y, Zhou F, Wang N, Xi H, et al. Circulating exosomal gastric cancer-associated long noncoding RNA1 as a biomarker for early detection and monitoring progression of gastric cancer: a multiphase study. JAMA Surg. 2020;155:572–579. doi: 10.1001/jamasurg.2020.1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Shao Y, Tao X, Lu R, Zhang H, Ge J, Xiao B, et al. Hsa_circ_0065149 is an indicator for early gastric cancer screening and prognosis prediction. Pathol Oncol Res. 2020;26:1475–1482. doi: 10.1007/s12253-019-00716-y. [DOI] [PubMed] [Google Scholar]
- 74.Zheng P, Gao H, Xie X, Lu P. Plasma exosomal hsa_circ_0015286 as a potential diagnostic and prognostic biomarker for gastric cancer. Pathol Oncol Res. 2022;28:1610446. doi: 10.3389/pore.2022.1610446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Aikou S, Ohmoto Y, Gunji T, Matsuhashi N, Ohtsu H, Miura H, et al. Tests for serum levels of trefoil factor family proteins can improve gastric cancer screening. Gastroenterology. 2011;141:837–845.e1. doi: 10.1053/j.gastro.2011.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Tong W, Ye F, He L, Cui L, Cui M, Hu Y, et al. Serum biomarker panels for diagnosis of gastric cancer. Onco Targets Ther. 2016;9:2455–2463. doi: 10.2147/OTT.S86139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Choi B, Lee HJ, Min J, Choe HN, Choi YS, Son YG, et al. Plasma expression of the intestinal metaplasia markers CDH17 and TFF3 in patients with gastric cancer. Cancer Biomark. 2017;19:231–239. doi: 10.3233/CBM-160147. [DOI] [PubMed] [Google Scholar]
- 78.Zhu Y, Hu Y, Zhu X, Zhang J, Yuwen D, Wei X, et al. Plasma thioredoxin reductase: a potential diagnostic biomarker for gastric cancer. Carcinogenesis. 2022;43:736–745. doi: 10.1093/carcin/bgac052. [DOI] [PubMed] [Google Scholar]
- 79.Park SH, Kang MJ, Yun EH, Jung KW. Epidemiology of gastric cancer in Korea: trends in incidence and survival based on Korea Central Cancer Registry data (1999-2019) J Gastric Cancer. 2022;22:160–168. doi: 10.5230/jgc.2022.22.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Feng F, Tian Y, Xu G, Liu Z, Liu S, Zheng G, et al. Diagnostic and prognostic value of CEA, CA19-9, AFP and CA125 for early gastric cancer. BMC Cancer. 2017;17:737. doi: 10.1186/s12885-017-3738-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zeng CD, Jin CC, Gao C, Xiao AT, Tong YX, Zhang S. Preoperative folate receptor-positive circulating tumor cells are associated with occult peritoneal metastasis and early recurrence in gastric cancer patients: a prospective cohort study. Front Oncol. 2022;12:769203. doi: 10.3389/fonc.2022.769203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ko K, Kananazawa Y, Yamada T, Kakinuma D, Matsuno K, Ando F, et al. Methylation status and long-fragment cell-free DNA are prognostic biomarkers for gastric cancer. Cancer Med. 2021;10:2003–2012. doi: 10.1002/cam4.3755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kim YW, Kim YH, Song Y, Kim HS, Sim HW, Poojan S, et al. Monitoring circulating tumor DNA by analyzing personalized cancer-specific rearrangements to detect recurrence in gastric cancer. Exp Mol Med. 2019;51:1–10. doi: 10.1038/s12276-019-0292-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Maron SB, Chase LM, Lomnicki S, Kochanny S, Moore KL, Joshi SS, et al. Circulating tumor DNA sequencing analysis of gastroesophageal adenocarcinoma. Clin Cancer Res. 2019;25:7098–7112. doi: 10.1158/1078-0432.CCR-19-1704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Yang J, Gong Y, Lam VK, Shi Y, Guan Y, Zhang Y, et al. Deep sequencing of circulating tumor DNA detects molecular residual disease and predicts recurrence in gastric cancer. Cell Death Dis. 2020;11:346. doi: 10.1038/s41419-020-2531-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Openshaw MR, Suwaidan AA, Ottolini B, Fernandez-Garcia D, Richards CJ, Page K, et al. Longitudinal monitoring of circulating tumour DNA improves prognostication and relapse detection in gastroesophageal adenocarcinoma. Br J Cancer. 2020;123:1271–1279. doi: 10.1038/s41416-020-1002-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yang C, Zou K, Yuan Z, Guo T, Xiong B. Prognostic value of circulating tumor cells detected with the CellSearch System in patients with gastric cancer: evidence from a meta-analysis. Onco Targets Ther. 2018;11:1013–1023. doi: 10.2147/OTT.S154114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ito H, Sato J, Tsujino Y, Yamaguchi N, Kimura S, Gohda K, et al. Long-term prognostic impact of circulating tumour cells in gastric cancer patients. World J Gastroenterol. 2016;22:10232–10241. doi: 10.3748/wjg.v22.i46.10232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Pernot S, Badoual C, Terme M, Castan F, Cazes A, Bouche O, et al. Dynamic evaluation of circulating tumour cells in patients with advanced gastric and oesogastric junction adenocarcinoma: Prognostic value and early assessment of therapeutic effects. Eur J Cancer. 2017;79:15–22. doi: 10.1016/j.ejca.2017.03.036. [DOI] [PubMed] [Google Scholar]
- 90.Ning D, Cui K, Liu M, Ou Y, Wang Z, Zou B, et al. Comparison of CellSearch and circulating tumor cells (CTC)-biopsy systems in detecting peripheral blood circulating tumor cells in patients with gastric cancer. Med Sci Monit. 2021;27:e926565. doi: 10.12659/MSM.926565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Szczepanik A, Sierzega M, Drabik G, Pituch-Noworolska A, Kołodziejczyk P, Zembala M. CD44+ cytokeratin-positive tumor cells in blood and bone marrow are associated with poor prognosis of patients with gastric cancer. Gastric Cancer. 2019;22:264–272. doi: 10.1007/s10120-018-0858-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Liu M, Wang R, Sun X, Liu Y, Wang Z, Yan J, et al. Prognostic significance of PD-L1 expression on cell-surface vimentin-positive circulating tumor cells in gastric cancer patients. Mol Oncol. 2020;14:865–881. doi: 10.1002/1878-0261.12643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Jhi JH, Kim GH, Park SJ, Kim DU, Lee MW, Lee BE, et al. Circulating tumor cells and TWIST expression in patients with metastatic gastric cancer: a preliminary study. J Clin Med. 2021;10:4481. doi: 10.3390/jcm10194481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.He Q, Li G, Ji X, Ma L, Wang X, Li Y, et al. Impact of the immune cell population in peripheral blood on response and survival in patients receiving neoadjuvant chemotherapy for advanced gastric cancer. Tumour Biol. 2017;39:1010428317697571. doi: 10.1177/1010428317697571. [DOI] [PubMed] [Google Scholar]
- 95.Ma J, Li J, He N, Qian M, Lu Y, Wang X, et al. Identification and validation of a novel survival prediction model based on the T-cell phenotype in the tumor immune microenvironment and peripheral blood for gastric cancer prognosis. J Transl Med. 2023;21:73. doi: 10.1186/s12967-023-03922-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Shin K, Kim J, Park SJ, Kim H, Lee MA, Kim O, et al. Early increase in circulating PD-1+CD8+ T cells predicts favorable survival in patients with advanced gastric cancer receiving chemotherapy. Cancers (Basel) 2023;15:3955. doi: 10.3390/cancers15153955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Fang WL, Lan YT, Huang KH, Liu CA, Hung YP, Lin CH, et al. Clinical significance of circulating plasma DNA in gastric cancer. Int J Cancer. 2016;138:2974–2983. doi: 10.1002/ijc.30018. [DOI] [PubMed] [Google Scholar]
- 98.Karamitrousis EI, Balgkouranidou I, Xenidis N, Amarantidis K, Biziota E, Koukaki T, et al. Prognostic role of RASSF1A, SOX17 and Wif-1 promoter methylation status in cell-free DNA of advanced gastric cancer patients. Technol Cancer Res Treat. 2021;20:1533033820973279. doi: 10.1177/1533033820973279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Li J, Li Z, Ding Y, Xu Y, Zhu X, Cao N, et al. TP53 mutation and MET amplification in circulating tumor DNA analysis predict disease progression in patients with advanced gastric cancer. PeerJ. 2021;9:e11146. doi: 10.7717/peerj.11146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Yan H, Chen W, Ge K, Mao X, Li X, Liu W, et al. Value of plasma methylated SFRP2 in prognosis of gastric cancer. Dig Dis Sci. 2021;66:3854–3861. doi: 10.1007/s10620-020-06710-8. [DOI] [PubMed] [Google Scholar]
- 101.Komatsu S, Imamura T, Kiuchi J, Takashima Y, Kamiya H, Ohashi T, et al. Depletion of tumor suppressor miRNA-148a in plasma relates to tumor progression and poor outcomes in gastric cancer. Am J Cancer Res. 2021;11:6133–6146. [PMC free article] [PubMed] [Google Scholar]
- 102.Matsusaka S, Chìn K, Ogura M, Suenaga M, Shinozaki E, Mishima Y, et al. Circulating tumor cells as a surrogate marker for determining response to chemotherapy in patients with advanced gastric cancer. Cancer Sci. 2010;101:1067–1071. doi: 10.1111/j.1349-7006.2010.01492.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Li Y, Zhang X, Liu D, Gong J, Wang DD, Li S, et al. Evolutionary expression of HER2 conferred by chromosome aneuploidy on circulating gastric cancer cells contributes to developing targeted and chemotherapeutic resistance. Clin Cancer Res. 2018;24:5261–5271. doi: 10.1158/1078-0432.CCR-18-1205. [DOI] [PubMed] [Google Scholar]
- 104.Cheng B, Tong G, Wu X, Cai W, Li Z, Tong Z, et al. Enumeration and characterization of circulating tumor cells and its application in advanced gastric cancer. Onco Targets Ther. 2019;12:7887–7896. doi: 10.2147/OTT.S223222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Negishi R, Yamakawa H, Kobayashi T, Horikawa M, Shimoyama T, Koizumi F, et al. Transcriptomic profiling of single circulating tumor cells provides insight into human metastatic gastric cancer. Commun Biol. 2022;5:20. doi: 10.1038/s42003-021-02937-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nose Y, Saito T, Yamamoto K, Yamashita K, Tanaka K, Yamamoto K, et al. The tissue-resident marker CD103 on peripheral blood T cells predicts responses to anti-PD-1 therapy in gastric cancer. Cancer Immunol Immunother. 2023;72:169–181. doi: 10.1007/s00262-022-03240-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Willis J, Lefterova MI, Artyomenko A, Kasi PM, Nakamura Y, Mody K, et al. Validation of microsatellite instability detection using a comprehensive plasma-based genotyping panel. Clin Cancer Res. 2019;25:7035–7045. doi: 10.1158/1078-0432.CCR-19-1324. [DOI] [PubMed] [Google Scholar]
- 108.Zhong Y, Fan Q, Zhou Z, Wang Y, He K, Lu J. Plasma cfDNA as a potential biomarker to evaluate the efficacy of chemotherapy in gastric cancer. Cancer Manag Res. 2020;12:3099–3106. doi: 10.2147/CMAR.S243320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Normando SR, Delgado PO, Rodrigues AK, David Filho WJ, Fonseca FL, Cruz FJ, et al. Circulating free plasma tumor DNA in patients with advanced gastric cancer receiving systemic chemotherapy. BMC Clin Pathol. 2018;18:12. doi: 10.1186/s12907-018-0079-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kim ST, Cristescu R, Bass AJ, Kim KM, Odegaard JI, Kim K, et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med. 2018;24:1449–1458. doi: 10.1038/s41591-018-0101-z. [DOI] [PubMed] [Google Scholar]
- 111.Kato S, Okamura R, Baumgartner JM, Patel H, Leichman L, Kelly K, et al. Analysis of circulating tumor DNA and clinical correlates in patients with esophageal, gastroesophageal junction, and gastric adenocarcinoma. Clin Cancer Res. 2018;24:6248–6256. doi: 10.1158/1078-0432.CCR-18-1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Chen Z, Zhang C, Zhang M, Li B, Niu Y, Chen L, et al. Chromosomal instability of circulating tumor DNA reflect therapeutic responses in advanced gastric cancer. Cell Death Dis. 2019;10:697. doi: 10.1038/s41419-019-1907-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Wang Y, Zhao C, Chang L, Jia R, Liu R, Zhang Y, et al. Circulating tumor DNA analyses predict progressive disease and indicate trastuzumab-resistant mechanism in advanced gastric cancer. EBioMedicine. 2019;43:261–269. doi: 10.1016/j.ebiom.2019.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wang DS, Liu ZX, Lu YX, Bao H, Wu X, Zeng ZL, et al. Liquid biopsies to track trastuzumab resistance in metastatic HER2-positive gastric cancer. Gut. 2019;68:1152–1161. doi: 10.1136/gutjnl-2018-316522. [DOI] [PubMed] [Google Scholar]
- 115.Li F, Yoshizawa JM, Kim KM, Kanjanapangka J, Grogan TR, Wang X, et al. Discovery and validation of salivary extracellular RNA biomarkers for noninvasive detection of gastric cancer. Clin Chem. 2018;64:1513–1521. doi: 10.1373/clinchem.2018.290569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Xu F, Jiang M. Evaluation of predictive role of carcinoembryonic antigen and salivary mRNA biomarkers in gastric cancer detection. Medicine (Baltimore) 2020;99:e20419. doi: 10.1097/MD.0000000000020419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Xiao H, Zhang Y, Kim Y, Kim S, Kim JJ, Kim KM, et al. Differential proteomic analysis of human saliva using tandem mass tags quantification for gastric cancer detection. Sci Rep. 2016;6:22165. doi: 10.1038/srep22165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Shu J, Yu H, Li X, Zhang D, Liu X, Du H, et al. Salivary glycopatterns as potential biomarkers for diagnosis of gastric cancer. Oncotarget. 2017;8:35718–35727. doi: 10.18632/oncotarget.16082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Huang K, Gao X, Wu L, Yan B, Wang Z, Zhang X, et al. Salivary microbiota for gastric cancer prediction: an exploratory study. Front Cell Infect Microbiol. 2021;11:640309. doi: 10.3389/fcimb.2021.640309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yu X, Luo L, Wu Y, Yu X, Liu Y, Yu X, et al. Gastric juice miR-129 as a potential biomarker for screening gastric cancer. Med Oncol. 2013;30:365. doi: 10.1007/s12032-012-0365-y. [DOI] [PubMed] [Google Scholar]
- 121.Cui L, Zhang X, Ye G, Zheng T, Song H, Deng H, et al. Gastric juice MicroRNAs as potential biomarkers for the screening of gastric cancer. Cancer. 2013;119:1618–1626. doi: 10.1002/cncr.27903. [DOI] [PubMed] [Google Scholar]
- 122.Shao J, Fang PH, He B, Guo LL, Shi MY, Zhu Y, et al. Downregulated MicroRNA-133a in gastric juice as a clinicopathological biomarker for gastric cancer screening. Asian Pac J Cancer Prev. 2016;17:2719–2722. [PubMed] [Google Scholar]
- 123.Shao Y, Ye M, Jiang X, Sun W, Ding X, Liu Z, et al. Gastric juice long noncoding RNA used as a tumor marker for screening gastric cancer. Cancer. 2014;120:3320–3328. doi: 10.1002/cncr.28882. [DOI] [PubMed] [Google Scholar]
- 124.Shao Y, Ye M, Li Q, Sun W, Ye G, Zhang X, et al. LncRNA-RMRP promotes carcinogenesis by acting as a miR-206 sponge and is used as a novel biomarker for gastric cancer. Oncotarget. 2016;7:37812–37824. doi: 10.18632/oncotarget.9336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Yang Y, Shao Y, Zhu M, Li Q, Yang F, Lu X, et al. Using gastric juice lncRNA-ABHD11-AS1 as a novel type of biomarker in the screening of gastric cancer. Tumour Biol. 2016;37:1183–1188. doi: 10.1007/s13277-015-3903-3. [DOI] [PubMed] [Google Scholar]
- 126.Zhou X, Liu J, Meng A, Zhang L, Wang M, Fan H, et al. Gastric juice piR-1245: a promising prognostic biomarker for gastric cancer. J Clin Lab Anal. 2020;34:e23131. doi: 10.1002/jcla.23131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Yamamoto H, Watanabe Y, Oikawa R, Morita R, Yoshida Y, Maehata T, et al. BARHL2 methylation using gastric wash DNA or gastric juice exosomal DNA is a useful marker for early detection of gastric cancer in an H. pylori-independent manner. Clin Transl Gastroenterol. 2016;7:e184. doi: 10.1038/ctg.2016.40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chen Q, Hu Y, Fang Z, Ye M, Li J, Zhang S, et al. Elevated levels of oxidative nucleic acid modification markers in urine from gastric cancer patients: quantitative analysis by ultra performance liquid chromatography-tandem mass spectrometry. Front Chem. 2020;8:606495. doi: 10.3389/fchem.2020.606495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Iwasaki H, Shimura T, Yamada T, Okuda Y, Natsume M, Kitagawa M, et al. A novel urinary microRNA biomarker panel for detecting gastric cancer. J Gastroenterol. 2019;54:1061–1069. doi: 10.1007/s00535-019-01601-w. [DOI] [PubMed] [Google Scholar]
- 130.Shimura T, Dagher A, Sachdev M, Ebi M, Yamada T, Yamada T, et al. Urinary ADAM12 and MMP-9/NGAL complex detect the presence of gastric cancer. Cancer Prev Res (Phila) 2015;8:240–248. doi: 10.1158/1940-6207.CAPR-14-0229. [DOI] [PubMed] [Google Scholar]
- 131.Dong X, Wang G, Zhang G, Ni Z, Suo J, Cui J, et al. The endothelial lipase protein is promising urinary biomarker for diagnosis of gastric cancer. Diagn Pathol. 2013;8:45. doi: 10.1186/1746-1596-8-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Shimura T, Dayde D, Wang H, Okuda Y, Iwasaki H, Ebi M, et al. Novel urinary protein biomarker panel for early diagnosis of gastric cancer. Br J Cancer. 2020;123:1656–1664. doi: 10.1038/s41416-020-01063-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Wu J, Zhang C, Xu S, Xiang C, Wang R, Yang D, et al. Fecal microbiome alteration may be a potential marker for gastric cancer. Dis Markers. 2020;2020:3461315. doi: 10.1155/2020/3461315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Liu S, Dai J, Lan X, Fan B, Dong T, Zhang Y, et al. Intestinal bacteria are potential biomarkers and therapeutic targets for gastric cancer. Microb Pathog. 2021;151:104747. doi: 10.1016/j.micpath.2021.104747. [DOI] [PubMed] [Google Scholar]
- 135.Okabe H, Tsunoda S, Hosogi H, Hisamori S, Tanaka E, Tanaka S, et al. Circulating tumor cells as an independent predictor of survival in advanced gastric cancer. Ann Surg Oncol. 2015;22:3954–3961. doi: 10.1245/s10434-015-4483-6. [DOI] [PubMed] [Google Scholar]
- 136.Takahashi K, Kurashina K, Yamaguchi H, Kanamaru R, Ohzawa H, Miyato H, et al. Altered intraperitoneal immune microenvironment in patients with peritoneal metastases from gastric cancer. Front Immunol. 2022;13:969468. doi: 10.3389/fimmu.2022.969468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hu XY, Ling ZN, Hong LL, Yu QM, Li P, Ling ZQ. Circulating methylated THBS1 DNAs as a novel marker for predicting peritoneal dissemination in gastric cancer. J Clin Lab Anal. 2021;35:e23936. doi: 10.1002/jcla.23936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Hiraki M, Kitajima Y, Koga Y, Tanaka T, Nakamura J, Hashiguchi K, et al. Aberrant gene methylation is a biomarker for the detection of cancer cells in peritoneal wash samples from advanced gastric cancer patients. Ann Surg Oncol. 2011;18:3013–3019. doi: 10.1245/s10434-011-1636-0. [DOI] [PubMed] [Google Scholar]
- 139.Han J, Lv P, Yu JL, Wu YC, Zhu X, Hong LL, et al. Circulating methylated MINT2 promoter DNA is a potential poor prognostic factor in gastric cancer. Dig Dis Sci. 2014;59:1160–1168. doi: 10.1007/s10620-013-3007-0. [DOI] [PubMed] [Google Scholar]
- 140.Tokuhisa M, Ichikawa Y, Kosaka N, Ochiya T, Yashiro M, Hirakawa K, et al. Exosomal miRNAs from peritoneum lavage fluid as potential prognostic biomarkers of peritoneal metastasis in gastric cancer. PLoS One. 2015;10:e0130472. doi: 10.1371/journal.pone.0130472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Ohzawa H, Saito A, Kumagai Y, Kimura Y, Yamaguchi H, Hosoya Y, et al. Reduced expression of exosomal miR-29s in peritoneal fluid is a useful predictor of peritoneal recurrence after curative resection of gastric cancer with serosal involvement. Oncol Rep. 2020;43:1081–1088. doi: 10.3892/or.2020.7505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Yang J, Cao W, Xing E. Levels and significance of tumor markers and cytokines in serum and peritoneal lavage fluid of patients with peritoneal metastasis of gastric cancer. BioMed Res Int. 2022;2022:9528444. doi: 10.1155/2022/9528444. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 143.Park HS, Kwon WS, Park S, Jo E, Lim SJ, Lee CK, et al. Comprehensive immune profiling and immune-monitoring using body fluid of patients with metastatic gastric cancer. J Immunother Cancer. 2019;7:268. doi: 10.1186/s40425-019-0708-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Tanaka Y, Chiwaki F, Kojima S, Kawazu M, Komatsu M, Ueno T, et al. Multi-omic profiling of peritoneal metastases in gastric cancer identifies molecular subtypes and therapeutic vulnerabilities. Nat Can. 2021;2:962–977. doi: 10.1038/s43018-021-00240-6. [DOI] [PubMed] [Google Scholar]
- 145.Wang R, Dang M, Harada K, Han G, Wang F, Pool Pizzi M, et al. Single-cell dissection of intratumoral heterogeneity and lineage diversity in metastatic gastric adenocarcinoma. Nat Med. 2021;27:141–151. doi: 10.1038/s41591-020-1125-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Yun J, Han SB, Kim HJ, Go SI, Lee WS, Bae WK, et al. Exosomal miR-181b-5p downregulation in ascites serves as a potential diagnostic biomarker for gastric cancer-associated malignant ascites. J Gastric Cancer. 2019;19:301–314. doi: 10.5230/jgc.2019.19.e27. [DOI] [PMC free article] [PubMed] [Google Scholar]


