Open Access

The diagnostic value of circulating microRNAs as a biomarker for gastric cancer: A meta‑analysis

  • Authors:
    • Hui Wei
    • Ke Pu
    • Xiao‑Guang Liu
    • Bo‑Xuan Li
    • Heng‑Shuo Zhang
    • Huan Wang
    • Hao Wang
    • Wei‑Ming Sun
    • Yu‑Ping Wang
  • View Affiliations

  • Published online on: October 11, 2018     https://doi.org/10.3892/or.2018.6782
  • Pages: 87-102
  • Copyright: © Wei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Recently, cancer research microRNA studies have drawn great attention. However, the results of these studies have been inconsistent and variable regarding the availability of circulating miRNAs in gastric cancer (GC) diagnosis. Thus, results should be interpreted cautiously. The purpose of the present study was to assess the diagnostic performance of circulating miRNAs in GC diagnosis. We conducted a systematic and comprehensive approach for the inclusion of studies. The sensitivity, specificity, and diagnostic odds ratio were pooled with random effects models, and a summary of receiver operator characteristic (SROC) curves were plotted. The potential heterogeneity was assessed with Q test and I2 statistics. Subgroup analyses and meta‑regressions further investigated the sources of heterogeneity. A total of 77 studies from 48 articles were eligible for the meta‑analysis. The results revealed a sensitivity of 0.76, a specificity of 0.81, and an AUC of 0.86 for gastric cancer diagnosis with circulating miRNAs. In addition, subgroup analyses indicated that multiple miRNAs assays, non‑microarray screening approaches, and serum‑based miRNA assays exhibited good diagnostic performance in contrast to a single miRNA assay, microarray expression profiling screening, and plasma‑based miRNA group analysis. The diagnostic ability of miRNAs in early stage I‑II groups and the high expression group were approximately similar to that in the stage I‑IV groups and the low expression group. For the circulating miRNAs, our meta‑analysis identified a combination of multiple miRNAs, non‑microarray chip screening, and serum‑based miRNA assays were associated with the most effective GC diagnostic performance. However, many unclear molecular mechanisms limited the accuracy of the diagnostic results, and should be interpreted with caution. Further large‑scale prospective studies are required for validating the diagnostic applicability of circulating miRNAs in gastric cancer patients.

Introduction

Gastric cancer (GC) is the sixth most common cancer and the fourth leading cause of cancer-related deaths worldwide (13). The incidence of GC, especially in the Asian region (China, Japan, and Korea), has markedly increased over the past three decades (4). Moreover, statistics of cancer research from the International Agency for Research on Cancer have revealed that the morbidity and mortality rates associated with GC in China accounted for 42.6 and 45.0% of the global rates in 2015, respectively (5). Consequently, the high incidence of GC can result in great health and economic burdens (6). The recent development of diagnostic and surgical techniques has greatly improved the prognosis of GC patients (7,8). Five-year survival rates for patients with early GC can reach 90%. Unfortunately, most patients, however, have already developed advanced GC at the time of diagnosis (9,10). For patients with advanced GC, several studies have revealed that the median survival time of patients with GC was only 6–9 months, with surgical treatment unable to prolong the patient survival (11,12). Even with expanded resection, lymph node clearance, and neoadjuvant chemotherapy, the incidence rates of postoperative local recurrence and distant metastasis are still high (13). Therefore, considering the high ratio of early GC patients who are cured, the key to reducing mortality rates and improving the prognosis for GC patients is early and accurate diagnosis.

To create a non-invasive, convenient, and low-priced diagnostic method, circulating biomarker detection methods have been widely used in clinics. GC serum tumor markers are currently used in clinical settings for the detection of GC. However, methods for the detection of carcinoembryonic antigen (CEA), carbohydrate antigen 199 (CA199), and carbohydrate antigen 724 (CA724) lack adequate sensitivity and specificity, which has precluded their widespread application in early diagnosis of GC (14). Therefore, it is necessary to investigate other potential biomarkers useful in identifying GC with high sensitivity and specificity. Recently, many researchers have been attracted to microRNAs (miRNAs) which have been stably detected in cell-free body fluids, such as plasma or serum. These miRNAs are called circulating miRNAs and are protected from degradation by ribonucleases in the blood. Additionally, the expression profile of miRNAs in GC patients usually exhibits exceptionally high expression of miRNAs in contrast to that in normal specimens (15).

miRNAs are a small, single-stranded non-coding RNAs that can regulate the expression of thousands of genes by inhibiting and degrading mRNAs during the translation process (16,17). miRNAs regulate various pathophysiological processes that involve cell proliferation, apoptosis, and differentiation. They also play an important role in tumorigenesis, especially neoplasm development, metastasis, angiogenesis, and immune responses (18).

Circulating miRNAs are considered to be novel potential biomarkers in the detection of many diseases, which were first demonstrated by Mitchell et al (19) for the detection of cancer. Subsequently, their roles have been demonstrated in numerous studies. Nevertheless, it is difficult to specify a suitable miRNA, and the results of miRNA expression analysis are inconsistent. Despite a large number of studies on circulating miRNAs, few relevant measures have been applied in the clinical setting. In the present study, in this meta-analysis, we systematically and comprehensively analyzed the diagnostic accuracy of circulating miRNAs in distinguishing gastric cancer patients from the significant heterogeneity of other factors.

Materials and methods

Literature search

Two authors independently searched online databases, PubMed, Embase, the Cochrane Library, Web of Science, and Springer to identify potentially eligible studies published before November 10, 2017. The keywords used for literature retrieval were (‘microRNA’ or ‘miRNA’ or ‘miR’) and (‘gastric cancer’ or ‘gastric carcinoma’ or ‘gastric tumor’ or ‘gastric neoplasm’) and (‘diagnostic’ or ‘diagnosis’ or ‘sensitivity and specificity’ or ‘ROC curve’) and (‘circulating’ or ‘plasma’ or ‘serum’ or ‘blood’). The citations in identified articles and in review articles were also examined. All publications identified by our search strategy were independently assessed by four reviewers. Any disagreement on a controversial study was resolved by discussion to consensus.

Literature selection

Eligible studies included in the present meta-analysis met the following criteria: i) the diagnosis of GC was made based on histopathological confirmation; ii) miRNA concentration in plasma, serum, or blood was detected before the patient received any treatment; iii) the study explored the correlation between miRNA expression levels and gastric cancer diagnosis; iv) the study included standard references for the GC diagnosis, including patients with benign diseases or healthy individuals as the control groups; and v) the study provided adequate or sufficient data for the calculation of 2×2 tables consisting of true positive (TP), false positive (FP), true negative (TN), and false negative (FN). In addition, exclusion criteria were: i) publications irrelevant to the detection values of circulating miRNAs for GCs; ii) review studies, editorials, case reports, and letters; iii) duplicate publications; and iv) unqualified data. All literature that satisfied the above criteria constituted qualified studies.

Data extraction and quality assessment

The following data characteristics were extracted for each eligible study by two reviewers (HW and KP), independently: i) basic characteristics of the included studies, including the first author, year of publication, country of publication, origin of the study population, sample type, study design, sample size, number of participants, and variables adjusted for the analysis, and ii) for diagnostic studies, the sensitivity, specificity, TP, FP, FN, and TN values of the diagnostic test results were considered.

The quality assessment of the studies included in the present study was performed by two authors independently using the Quality Assessment of Diagnostic Accuracy studies (QUADAS-2) (20) checklist in Rev Man 5.0 software (http://ims.cochrane.org/revman/download). This scale was composed of four domains consisting of patient selection, index test, reference standard, and flow and timing domain. Each signaling question was judged as ‘yes’, ‘no’, or ‘unclear’ and each study's risk of bias and concern for applicability was estimated as ‘high’, ‘low’, or ‘unclear’ except for the flow and timing domain, for which the applicability concern did not apply. An answer of ‘yes’ meant that the risk of bias could be judged as being low, whereas an answer of ‘no’ or ‘unclear’ meant that the risk of bias could be judged as being high. A third reviewer was consulted in the case of conflict and inconsistency was dealt with by a multilateral argument.

Statistical methods

To evaluate the diagnostic accuracy of miRNA assays for detecting GC, the sensitivity and specificity parameters were extracted from each study and the number of patients with TP, FP, TN, and FN results from each included article were extracted or calculated. Based on the 2×2 tables, meta-analyses were performed with Meta-Disc software version 1.4 (http://www.hrc.es/investigacion/metadisc_en.htm.) and STATA software version 12.0 (Stata Corporation, College Station, TX, USA) to evaluate the pooled statistics (95% CI) of sensitivity, specificity, positive and negative likelihood ratios (PLR and NLR) [PLR = sensitivity/(1 - specificity), NLR = (1 - sensitivity)/specificity], diagnostic odds ratio (DOR), and area under the summary receiver operating characteristic curves (AUSROC), with standard errors (SE) and Q index with SE for the test's performance using miRNA for GC detection. If sufficient information was not available, we recalculated these values on the basis of the sensitivity and specificity offered. Summary statistics revealed the diagnostic threshold effects that were analyzed by Spearman's correlation coefficient and P-value. If there was no significant threshold effect, the diagnostic accuracy was estimated by pooled statistics, whereas the diagnostic accuracy was evaluated only by AUSROC and Q indices rather than sensitivities, specificities, PLR, NLR, and DOR.

Positive and negative likelihood ratios describe the discriminatory properties of positive and negative test results, respectively (21). Likelihood ratios state how many times more likely particular test results are in patients with disease than in those without disease (22). Positive likelihood ratios above 10 and negative likelihood ratios below 0.1 have been noted as providing convincing diagnostic evidence, whereas PLR>5.0 and NLR<0.2 imply higher diagnostic evidence (23). DOR represented the positive odds of aberrant miRNA expression in patients with GC compared to the probability of the healthy control. AUSROC values of 0.5–0.7, 0.7–0.9, and 0.9–1.0 were applied to indicate low, moderate, and high diagnostic accuracy, respectively. A smaller Q index indicated a lower diagnostic accuracy.

Heterogeneity was determined by Cochran's Q statistic based on the Chi-square test and I2 statistics. I2 values of 0–40, 40–70, and 70–100% indicated low, moderate, and high variance, respectively (24). If moderate heterogeneity existed or different clinical characteristics were noted, the DerSimonian and Laird random-effects model was applied. Considerable heterogeneity was considered if I2>50% and/or P<0.05 (25,26). Sources of heterogeneity were explored by meta-regression analysis based on possible characteristics and a subsequent subgroup analysis was performed to identify potential covariates. Furthermore, subgroup and meta-regression analyses were performed to detect the extent of heterogeneity between studies. Publication bias was checked using Deeks' funnel plot analysis (24). All of the aforementioned statistical calculations were made with Meta-DiSc and STATA 12.0 software.

Results

Literature search

In Fig. 1, the flowchart for the selection of included articles is presented. Searching Pubmed, Embase, the Cochrane library, and Web of Science resulted in the inclusion of 531 articles. After a review of titles and abstracts, 156 publications were found to be irrelevant or duplicated. Next, we, intensively read the remaining studies, whereby 114 of these publications were removed for irrelevant content, 153 articles were eliminated owing to the study being unrelated to gastric cancer, and 26 studies were not considered as they were review letters and meta-analyses. After a more detailed evaluation, 34 studies were removed as they did not contain full text or had insufficient data for extraction. Finally, the selection process revealed 48 studies that were eligible for diagnostic analysis.

Study characteristics and quality assessment

The main characteristics of the 48 qualified articles included 77 microRNAs, of which one study was performed in Europe and 47 studies were undertaken in Asia. The evaluated studies included a total of 3,829 cases and 3,175 controls for the present meta-analysis. These are presented in Table I. In the tumor-node-metastasis (TNM) classification, 20 articles included patients in stages I–IV. The other 13 included patients in stages I–II, of which only one study involved patients in stage I, whereas the remainder of the eligible studies (n=15) did not mention the TNM stage. Analysis of data from the nine included studies that used miRNA microarray chips and revealed a number of miRNAs with altered expression, where candidate miRNAs were chosen via the training and validation design, whereas candidate miRNAs from the remaining articles (n=39) were chosen directly without microchip procession. Meanwhile, 42 of the 48 studies investigated the diagnostic value of a single miRNA used in GC detection, while only nine researched a set of miRNAs. Of the selected miRNAs, three were from the single miRNA studies. In terms of samples, circulating miRNAs from GC and healthy individuals were classified as serum (n=17), plasma (n=25), and peripheral blood (n=6). We also summarized miRNAs whose expression was upregulated in 33 studies and downregulated in 15 studies. One study (8) was excluded due to the unclear description about miRNA regulation, while another study (38) involved both upreguated and downregulated miRNAs. In particular, the expression of 58 miRNAs, including miR-21, was most frequently upregulated and that of 18 miRNAs (miR-26a and miR-199) was downregulated.

Table I.

Descriptive characteristics of the eligible studies.

Table I.

Descriptive characteristics of the eligible studies.

Case/control

AuthorYearRegionSample size (n)Mean age (case/control) (year)Male ratio (case/control) (%)Sample/methodsmiRNA profilingPatients TNM (I/II/III/IV)(Refs.)
Peng WZ2014China57/58NMNMSerum/qRT-PCRmiR-191NM(27)
Qiu X2016China280/28063.3/63.263.2/63.2Plasma/qRT-PCRmiR-26a65/61/135/16/3 (I/II/III/IV/missing)(28)
Tsujiura M2015Japan104/65NMNMPlasma/qRT-PCRmiR-18a66/14/15/9(29)
Zhang J2015China155/11156.6/47.471/63Plasma/qRT-PCRmiR-16-5p, 19b-3p33/26/59/25/12 (I/II/III/IV/unknown)(30)
Tsai MM2016Taiwan98/12664.1/66.458.2/47.6Plasma/qRT-PCRmiR-196a, 196b, and both30/12/38/18(31)
Su ZX2014China82/6569.0/71.258.5/60.0Plasma/qRT-PCRmiR-18aNM(32)
Valladares-Ayerbes M2012Spain52/1565.9/65.381/80Blood/qRT-PCRmiR-200c9/12/31 (I–II/III/IV)(33)
Zhu X2017China114/95NM62.0/64.1Serum/qRT-PCRmiR-14535/79 (I+II/III+IV)(34)
Zhuang K2016China138/50NM61.6/NMPlasma/qRT-PCRmiR-23bNM(35)
Hung PS2017Taiwan65/10867.7/56.172.3/62.0Plasma/qRT-PCRmiR-376c30/14/16/5(36)
Huang S2016China62/5958/57NMSerum/qRT-PCRmiR-21, 31, 92a, 181b, 203NM(8)
Le Q2017China41/41NM58.5/53.7Blood/qRT-PCRmiR-25NM(37)
Li BS2012China70/70NM70/63Plasma/qRT-PCRmiR-223, 21, 21812/11/36/11(38)
Li F2017China65/6554.1/56.276.92/76.92Plasma/RT-PCRmiR-106b, 93, 2529/36 (I+II/III+IV)(39)
Li M2017China51/51NM62.7/NMPlasma/qRT-PCRmiR-200cNM(40)
Hou X2015China80/8068.0/67.057.5/55Plasma/qRT-PCRmiR-106a45/35 (I+II/III+IV)(41)
Hou CG2016China150/150NM65.33/NMSerum/qRT-PCRmiR-20657/93 (I+II/III+IV)(42)
Zhang WH2012China20/2060.9/NM75/NMSerum/qRT-PCRmiR-3751/1/1/6/4/7 (Ib/II/IIIa/IIIb/IV/unknown)(43)
Song MY2012China68/68NMNMSerum/qRT-PCRmiR-221, 376c, 744NM(44)
Tsujiura M2010Japan69/30NMNMPlasma/qRT-PCR miR-106a/let-7a38/13/14/4(45)
Wang B2012China30/3958.0/46.073.3/23.1Serum/qRT-PCRmiR-2111/19 (I+II/III+IV)(46)
Zhu C2014China48/10256.6/54.072.9/70.6Plasma/qRT-PCRmiR-16, 25, 92a, 451, 486-5p, and combination
Liu R2011China82/6460.2/60.084.0/78.0Serum/qRT-PCRCombined (miR-1, 20a, 27a, 34a, 423-5p)29/56/48/23/8 (I/II/III/IV/unknown)(48)
Konishi H2012Japan56/30NMNMPlasma/qRT-PCRmiR-451, 486NM(49)
Liu H2012China40/4156.0/58.065.6/65.6Plasma/qRT-PCRmiR-3784/12/11/13(50)
Liu X2016Hong Kong80/7067.0/56.066.9/59.0Plasma/qRT-PCRmiR-94020/10/28/22(51)
Hu Y2016China137/79NM53.3/NMSerum/qRT-PCRmiR-133a58/79 (I+II/III+IV)(52)
Sun Y2017China76/26NM96.7/61.5Serum/qRT-PCRmiR-18315/10/30/21(53)
Li B2017China116/85NM66.38/NMPlasma/qRT-PCRmiR-32068/48 (I+II/III+IV)(54)
Wu J2015China90/90NM48.9/NMSerum and PBMCs/qRT-PCRmiR-42132/21/9/28(55)
Zeng Q2014China40/36NM70/NMSerum/qRT-PCRmiR-17, 106b and both9/31 (I/II)/(III/IV)(56)
Zhou H2012China40/17NM75/NMBlood/qRT-PCRmiR-4218/13/4/5/10 (I/II/III/IV/unknown)(57)
Zhou X2015China32/18NM65.6/61.1Plasma/qRT-PCRMultiple (miR-185, 20a, 210, 25, 92b)6/5/17/4(58)
Wu J2015China50/50NM48/NMSerum and PBMCs/qRT-PCRmiR-219/11/10/18/2 (I/II/III/IV/unknown)(59)
Wu D2017China32/32NMNMSerum samples/qRT-PCRmiR-503NM(60)
Wang H2014China50/47NM54/NMSerum/qRT-PCRmiR-223, 16, 10031/19 (I+II/III+IV)(61)
Shin VY2015Hong Kong108/96NMNMPlasma/qRT-PCRMultiple (miR-627, 629, 652)16/13/41/38(62)
Cui YJ2015China46/40NM67.4/55Plasma/RFQ-PCRmiR-27b-3p8/15/12/11(63)
Fu Z2014China114/56NM47.36/NMPlasma/qRT-PCRmiR-22217/25/34/38(64)
Liu H2017China137/14554.3/53.662/64.1Plasma/qRT-PCRmiR-21743/94 (I+II/III+IV)(65)
Zhou H2010China41/27NMNMPMNCs/qRT-PCRmiR-106a, 17, bothNM(66)
Zhou X2016China45/45NMNMSerum/qRT-PCRmiR-223NM(67)
Zheng Y2011China52/20NMNMBlood/qRT-PCRmiR-21NM(68)
Li H2017China75/38NMNMBlood/qRT-PCRmiR-17-3p, 17-5p, 18a-5p, 19a-3p, 20a-5p, 92a-3pNM(69)
Park JL2015Korea35/3551.8/48.951.4/51.4Plasma/qRT-PCRmiR-27aNM(70)
Li C2013China80/7056.7/58.968.8/64.3Plasma/qRT-PCRmiR-199a-3p69/7/4 (Ia/Ib/IIa)(71)
Li C2013China180/13058.1/58.968.9/65.4Plasma/qRT-PCRmiR-199a-3p40/29/93/18(72)
Liu S2017China96/40NMNMSerum/qRT-PCRmiR-14447/49 (I+II/III+IV)(73)

[i] TP, true positive; FP, false positive; FN, false negative; TN, true negative; SEN, sensitivity; SPE, specificity; AUSROC, area under the summary ROC curve; NM, not mentioned.

The quality of the eligible studies which were assessed, based on the QUADAS-2 criteria, was independently appraised by reviewers and is reported in Fig. 2A and B. In total, only five studies were valued as being low risk for bias and applicability concerns. The remaining studies were estimated as suboptimal for unclear risk in areas including index test, reference standard, flow, and timing. Most of the studies were identified as having a potential bias risk for patient selection and reference standard.

Diagnostic accuracy of circulating miRNA in GC

First, Meta-DiSc software version 1.4 was used to analyze the data. The heterogeneity test found that Q test and I2 values of DOR were 233.49 and 67.5%, respectively (P=0.0000) (Fig. 3A). Next, we generated forest plots of sensitivity and specificity, both of which did not display a straight line distribution and the Cochran-Q values were 369.52 and 555.63 (Fig. 3B and C), respectively, which reflected substantial heterogeneity (79.4 and 86.3%, respectively) among these studies. Random-effects models were then selected to re-analyze the data and the diagnostic threshold was analyzed. The Spearman's correlation coefficient was 0.253 (P=0.026), illustrating that the significant heterogeneity was partially caused by the diagnostic threshold. In addition, this may be caused by discrepancies in the study approaches, specimen type, endogenous reference, or total sample size. Thus, we could not calculate the statistical outcomes indirectly by neglecting the different factors and owing to the high heterogeneity. The data could not simply be pooled and was only suitable for subgroup analyses for illustrating heterogeneity.

Covariates and subgroup analysis

After stratification in accordance with the five pre-specified covariates, including patients' stage of GC (early TNM stage I–II vs. all TNM stages I–IV), miRNA profiling (single miRNA vs. multiple miRNAs), specimen types (plasma vs. serum vs. blood), miRNA screening approaches (microarray processing vs non-microarray processing), and aberrant expression (upregulation vs. down-regulation). we next assessed their impact on sensitivity or/and specificity as shown in Table II and Fig. 4. Comparing different TNM stages of GC patients with altered expression of circulating miRNAs, the results revealed that the diagnostic accuracy of miRNA detection during early stages I–II (sensitivity, 0.76; specificity, 0.85; PLR, 5.20; NLR, 0.28; DOR, 17.63; and AUC, 0.87) was similar to that during all stages I–IV (sensitivity, 0.80; specificity, 0.83; PLR, 4.70; NLR, 0.24; DOR, 18.01; and AUC, 0.87) and non-description stages (sensitivity, 0.76; specificity, 0.83; PLR, 4.40; NLR, 0.29; DOR, 12.18; and AUC, 0.85) with respect to all parameters except for the slight disparity in DOR. These results indicated that these biomarkers could not differentiate early GC from other later TNM stages in accordance with the diagnostic value.

Table II.

Summary estimates of diagnostic criteria and their 95% confidence intervals.

Table II.

Summary estimates of diagnostic criteria and their 95% confidence intervals.

SubgroupN (miR)SEN (95% CI)SPE (95% CI)PLR (95% CI)NLR (95% CI)DOR (95% CI)I2 (%)AUC (95% CI)
TNM stage
  TNM (I–II)240.76 (0.72–0.79)0.85 (0.81–0.89)5.20 (4.16–6.52)0.28 (0.25–0.32)17.63 (13.82–22.48)41.20.87 (0.84–0.89)
  TNM (I–IV)270.80 (0.75–0.84)0.83 (0.76–0.88)4.70 (3.32–6.66)0.24 (0.20–0.29)18.01 (12.56–25.81)74.50.87 (0.84–0.90)
  Unclassified TNM stage260.76 (0.71–0.80)0.83 (0.76–0.88)4.40 (3.12–6.21)0.29 (0.24–0.36)12.18 (8.40–17.68)67.50.85 (0.82–0.88)
miRNA screening
  Mircroarray200.78 (0.72–0.83)0.81 (0.72–0.87)4.04 (2.81–5.80)0.28 (0.22–0.35)12.35 (8.63–17.68)68.70.85 (0.82–0.88)
  Unrelated microarray570.77 (0.74–0.80)0.85 (0.81–0.88)5.01 (4.11–6.12)0.27 (0.24–0.30)17.20 (13.66–21.64)66.70.87 (0.84–0.90)
miRNA expression profiling
  Single miR680.77 (0.74–0.80)0.84 (0.80–0.87)4.69 (3.87–5.67)0.28 (0.25–0.31)17.00 (13.39–21.56)67.20.86 (0.83–0.89)
  Multiple miRs90.80 (0.75–0.84)0.85 (0.76–0.90)5.21 (3.22–8.43)0.23 (0.18–0.31)22.22 (11.03–44.77)68.20.87 (0.84–0.90)
Sample type
  Serum230.81 (0.78–0.84)0.83 (0.77–0.88)4.89 (3.55–6.72)0.23 (0.19–0.27)19.44 (13.33–28.36)65.80.88 (0.85–0.90)
  Plasma400.78 (0.74–0.81)0.84 (0.79–0.88)4.90 (3.74–6.43)0.27 (0.23–0.31)16.47 (12.62–21.49)72.20.86 (0.83–0.89)
  Peripheral blood140.68 (0.61–0.74)0.81 (0.75–0.86)3.62 (2.81–4.65)0.39 (0.33–0.47)8.82 (6.36–12.24)15.60.82 (0.79–0.85)
Altered miRNAa
  Upregulation580.76 (0.73–0.79)0.84 (0.80–0.87)4.75 (3.84–5.88)0.28 (0.25–0.32)15.18 (11.98–19.25)68.30.86 (0.82–0.89)
  Downregulation180.80 (0.75–0.84)0.81 (0.75–0.82)4.25 (3.22–5.61)0.25 (0.20–0.31)15.93 (11.61–21.86)59.80.87 (0.84–0.90)

a Huang S et al(8) was excluded due to the unclear description about miRNA regulation. SEN, sensitivity; SPE, specificity; NLR, negative likelihood ratio; PLR, positive likelihood ratio; DOR, diagnostic odds ratio; CI, confidence interval.

Subsequently, we focused on the different screening approaches, such as microarray processing vs. non-microarray processing, and applied these approaches to candidate miRNAs. The differences between the pooled estimates of DOR (Table II) between miRNAs originating from microarray and miRNAs selected directly implied that the diagnostic ability of the former was inferior to the latter. The AUSROC (Fig. 5) indicated that the diagnostic accuracy of miRNAs in microarray screening was slightly less than the non-microarray selection group. In addition, we conducted subgroup analyses based on miRNA profiling, including single miRNA and multiple miRNAs. In the subgroup analysis (Table II and Fig. 6), compared to that of single miRNA, the diagnostic ability of multiple miRNAs was better, with the sensitivity increasing from 0.77 to 0.80 and the specificity increasing from 0.84 to 0.85. AUC varied from 0.86 to 0.87 and the DOR value markedly increased from 17.00 to 22.22, whereas AUROC implied that there were parallel diagnostic accuracies between both.

Subgroup analysis of specimen types and aberrant expression were conducted to identify whether the candidate miRNAs could clearly discriminate GC sufficient diagnostic performance and accuracy. In the specimen type subgroup revealed in Table II and Fig. 7, the diagnostic sensitivity of miRNAs extracted from serum was 0.81 and the specificity was 0.83, with a pooled DOR of 19.44 and AUC of 0.88. The sensitivity and specificity of miRNAs from plasma-based studies were 0.78 and 0.84, respectively, with a summary DOR of 16.47 and AUC of 0.86. However, for the peripheral blood assay, sensitivity and specificity were 0.68 and 0.81, respectively, with a pooled DOR of 8.82 and AUC of 0.82, which indicated that the serum-based miRNA detection had a higher diagnostic value for GC than either the plasma or the peripheral blood-based assays. Moreover, further research was conducted to identify whether the upregulated miRNAs had better diagnostic accuracy than the downregulated miRNAs. Thus, the altered expression subgroup analyses for all miRNAs are shown in Table II and Fig. 8, the variable value in both changed slightly, according to the data of DOR (15.18 and 15.93) and AUC (0.86 and 0.87), which revealed that the diagnostic performance of miRNAs for GC detection in the high expression group was similar to that in the low expression group. From the above subgroup analysis that sought to determine the source of heterogeneity, the results indicated that the subgroups involving patients in the TNM (I–II) stage (I2=41.2%) using peripheral blood samples (I2=15.6%), contributed to moderate and mild heterogeneity, respectively. In addition, the decreasing trend in alteration implicitly suggested that the two factors may possibly be the source of heterogeneity. Nevertheless, further steps were taken to determine whether the aforementioned controversy led to the heterogeneity partly or entirely, which was confirmed by meta-regression analysis.

Meta-regression analysis

Based on the characteristics of the included studies, covariates, including TNM classification of GC, miRNA profiling, specimen types, miRNAs screening approaches, and aberrant expression status were applied to investigate inter-study heterogeneity using a meta-regression model shown in Table III (A-E). In the meta-regression analysis, sources of significant heterogeneity statistically indicated that the specimen type of the miRNA contributed significantly (P=0.0014), while the heterogeneity of results was not significantly influenced by the rest of the covariates. In accordance with the above results, the study sample type could be considered as a source of heterogeneity for GC detection in meta-regression.

Table III.

Meta regression to evaluate the inter-subgroup heterogeneity of miRNAs for the diagnosis of gastric cancer patients.

Table III.

Meta regression to evaluate the inter-subgroup heterogeneity of miRNAs for the diagnosis of gastric cancer patients.

A, Five covariates

VariablesCoeff.Sth. ErrP-valueRDOR(95% CI)
Cte.3.0590.43430.0000
S−0.3010.07350.0001
TNM stage0.1860.11190.10041.20(0.96;1.51)
Up/downregulation−0.0120.20470.95350.99(0.66;1.49)
miR screening−0.4780.20510.02260.62(0.41;0.93)
Sample type−0.4150.13610.00320.66(0.50;0.87)
Single/multiple0.2540.22990.27281.29(0.82;2.04)

Tau-squared estimate=0.2689 (convergence is achieved after 6 iterations). Restricted Maximum Likelihood estimation (REML). No. studies=48 containing 77 miRNAs. Filter OFF. Add 1/2 to all cells of the studies with zero. Cte, constant coefficient; S, statistic; RDOR, relative diagnostic odds ratio.

B, Four covariates

VariablesCoeff.Std. ErrP-valueRDOR(95% CI)

Cte.3.0480.41830.0000
S−0.3000.07300.0001
TNM stage0.1880.10780.08511.21(0.97;1.50)
miR screening−0.4790.20290.02090.62(0.41;0.93)
Sample type−0.4160.13290.00250.66(0.51;0.86)
Single/multiple0.2550.22720.26581.29(0.82;2.03)

Tau-squared estimate=0.2606 (convergence is achieved after 6 iterations). Restricted Maximum Likelihood estimation (REML). No. studies=48 containing 77 miRNAs. Filter OFF. Add 1/2 to all cells of the studies with zero. Cte, constant coefficient; S, Statistic; RDOR, relative diagnostic odds ratio.

C, Three covariates

VariablesCoeff.Std. ErrP-valueRDOR(95% CI)

Cte.3.3640.31560.0000
S−0.3280.06960.0000
TNM stage0.1760.10860.10851.19(0.96;1.48)
miR screening−0.4020.19320.04090.67(0.46;0.98)
Sample type−0.4280.13400.00210.65(0.50;0.85)

Tau-squared estimate=0.2727 (convergence is achieved after 6 iterations). Restricted Maximum Likelihood estimation (REML). No. studies=48 containing 77 miRNAs. Filter OFF. Add 1/2 to all cells of the studies with zero. Cte, constant coefficient; S, Statistic; RDOR, relative diagnostic odds ratio.

D, Two covariates

VariablesCoeff.Std. ErrP-valueRDOR(95% CI)

Cte.3.6300.27590.0000
S−0.3280.07060.0000
miR screening−0.3550.19440.07160.70(0.48;1.03)
Sample type−0.4760.13280.00060.62(0.48;0.81)

Tau-squared estimate=0.2899 (convergence is achieved after 6 iterations). Restricted Maximum Likelihood estimation (REML). No. studies=48 containing 77 miRNAs. Filter OFF. Add 1/2 to all cells of the studies with zero. Cte, constant coefficient; S, Statistic; RDOR, relative diagnostic odds ratio.

E, One covariate

VariablesCoeff.Std. ErrP-valueRDOR(95% CI)

Cte.3.4740.26610.0000
S−0.3350.07170.0000
Sample type−0.4450.13400.00140.64(0.49;0.84)
Tau-squared estimate=0.3116 (convergence is achieved after 6 iterations). Restricted Maximum Likelihood estimation (REML). No. studies=48 containing 77 miRNAs. Filter OFF. Add 1/2 to all cells of the studies with zero. Cte, constant coefficient; S, Statistic; RDOR, relative diagnostic odds ratio.
Publication bias

Deeks' funnel plot asymmetry test was applied to explore the publication bias of meta-analysis in diagnostic accuracy (24). The slope coefficient was associated with a P-value of 0.756 for GC detection (Fig. 9), suggesting a low likelihood of publication bias in our meta-analysis.

Discussion

GC is responsible for the highest number of cancer-related mortalities (74), and the egregious mortality of GC is immeasurably more acute than ever before, primarily since the majority of patients have a terminal disease at stage III or IV at the time of diagnosis (75). In addition, there are many investigated biomarkers, such as CEA and CA724, which lack sufficient sensitivity and specificity for early GC diagnosis (76), and universal screening tools, such as endoscopic examinations and biopsies, are invasive, unpleasant, and inconvenient, leading to potential errors in GC detection. Hence, ideal non-invasive biomarkers are urgently required to reinforce GC detection. At present, a large number of studies on the search for novel tumor biomarkers have revealed that miRNAs may play a pivotal role in cancer suppression, owing to the diverse miRNA expression levels that are observed between cancer patients and healthy controls (45,48,77,78). Subsequently, research has gravitated towards miRNAs as biological markers for tumor diagnosis. Nevertheless, the results of research on the use of miRNAs for gastrointestinal cancer detection are conflicting among different studies (33,38,44,45,47,48,79,80). To the best of our knowledge, several meta-analysis studies have been undertaken to determine the differentially expressed miRNAs in GC patients. Unfortunately, as a result of insufficient data or inconsistent results, abundant heterogeneity influenced the results. Furthermore, the accuracy of performance using the pooled results influenced interpretation. In the present meta-analysis, we included 48 studies involving miRNA expression profiling to systematically and comprehensively evaluate the potential diagnostic value of circulating miRNAs as diagnostic markers for GC. We considered different perspectives while avoiding statistical outcomes that included the absence of homogeneity.

The pooled outcomes of sensitivity, specificity, and AUC (0.76, 0.81, and 0.86, respectively) with the random effects model revealed that circulating miRNAs have better diagnostic value than CEA and CAA199 (AUC of 0.55 and 0.60, respectively) in distinguishing GC patients from control groups. Moreover, the DOR of circulating miRNAs for GC detection was 15.72, reflecting higher diagnostic performance as a combinative parameter of sensitivity and specificity. In fact, by pooling data in this manner, the diagnostic value would be inaccurate due to the significant heterogeneity and diagnostic threshold. Thus, we could not interpret the statistical outcomes blindly while neglecting high heterogeneity. Additionally, it was suitable to explore subgroup and regression analyses.

From the subgroup analysis, our results indicated that the non-microarray screening approach, multiple miRNA assay, and serum-based miRNA assay manifested a relatively higher diagnostic value and accuracy for GC than the single-miRNA, microarray profiling screening, and plasma-based miRNA groups. The altered expression of the single miRNA in serum or plasma fluctuated not only in GC but also in other tumors, infectious diseases, nonspecific inflammation, and acute injuries. In other words, single miRNAs lacked specificity in cancer detection. However, for multiple miRNAs with complex molecular mechanisms, such as a competing endogenous RNA (ceRNA) network intersecting at tumorigenesis (e.g., initiation and development of a severe neoplasm), the association may be valuable for early GC detection. Hence, studies highlighting individual cancer-specific miRNAs in serum or plasma usually arrived at unsatisfactory results.

Non-microarray screening and serum-based diagnostic tests yielded better outcomes than microarray screening pathways and plasma-based investigations of GC. However, the origin of source-related differences was still unclear. There are many complex factors that must be accounted for, such as lower homogeneity of included studies and a limited number of samples. Therefore, large-scale investigations and multiple center trials should be undertaken in the future to uncover the underlying mechanism of aberrant expression of miRNAs and to determine whether the source-related discrepancies truly exist or not.

Another finding was that the diagnostic value of miRNAs in early stages I–II and high expression groups were approximately similar to those in stages I–IV and lower expression groups in the detection of GC. In the GC microenvironment, a variety of differentiated tumor cells and cancer-associated cells, such as different types of immune cells and cells with different proliferative activity, lead to the transcriptome dysfunction during the tumorigenesis process due to inactivation of tumor suppressors and activation of proto-oncogenes. With respect to cancer, immune cells have the capacity to release exosomes that accompany cell migration, shuttling the ceRNA network into circulation (81), whereas the circulating tumor cells may express non-coding RNA under the control of an oncogene. Moreover, the overexpression of miRNAs may silence the mRNAs from tumor suppressor genes and downregulated miRNAs may facilitate the expression of oncogene mRNAs by binding the 3′-untranslated region of the target mRNA. In early and advanced GC, the aberrant and abundant expression of some miRNAs may be associated with this process. Moreover, a lack of specificity in the aforementioned may occur not only in GC but in many tumors, leading to various conclusions regarding GC discrimination. For instance, Liu et al (50) suggested that miRNA expression levels during earlier stages (I and II) were different from those during later stages (III and IV), exhibiting that this miRNA could be valuable for the early detection of GC. Evidently, we arrived at a paradox with our results conflicting with the conclusions of these authors. Therefore, it is difficult to distinguish which is a suitable and specific biomarker for GC. It would be beneficial to study individual miRNAs by determining their molecular mechanism rather than using the combination methodology.

The present study does have some limitations that must be addressed. First, methodologies for a precise uniform quantification of miRNAs face a lack of consistent criteria, limiting the comparisons made between studies that are conducted by different laboratories who have their own study design, use of miRNA chips, pathology type, localization of GC lesions, and different endogenous miRNA references. Second, there are some specific circulating miRNAs that are always prone to be selected by certain studies in determining the correlation between grade and stage of cancer. Consequently, a standardized protocol, which would be preferable, is required to abate bias. Moreover, the included studies in the present meta-analysis only distinguished the tumor patients from healthy controls, but other risk factors, such as chronic gastritis, infectious disease, genetic, ulcers, and diet, were not included and these may contribute to altered miRNA expression (4,82). Therefore, further accurate studies on the use of miRNAs for distinguishing cancer from other diseases are urgently needed.

In conclusion, our meta-analysis found that the combination of multiple miRNAs, non-microarray chip screening, and serum-based miRNA assays may present a better performance for the diagnosis of GC. However, many unclear molecular mechanisms hindered discovery for clear GC detection biomarkers. Therefore, the results should be interpreted cautiously given the uncertainty of the results. Further large-scale prospective studies are required to validate the potential applicability in human cancer diagnosis.

Acknowledgements

I would like to thank all authors that were involved in this study. In addition, we are appreciative of the support from Professors YPW and WMS.

Funding

This study was supported by the National Natural Science Foundation of China (81570783), the National Key Research and Development Program of China (2016YFC1302200), the Gansu Natural Science Foundation of China (1606RJZA136), the Health Industry Research Project of the Gansu Province (GWGL2014-01), the Scientific Research Project of Traditional Chinese Medicine Administration in Gansu Province (GZK-2017-48), the Open Fund of State Key Laboratory of Cancer Biology (CBSKL201718), and the Fundamental Research Funds for the Central Universities (lzujbky-2015-258).

Availability of data and materials

All data generated or analyzed in this study are included in this published article.

Authors' contributions

HuiW, KP, XGL, BXL, HSZ, HuanW, HaoW, WMS and YPW contributed to the conception and design of the study, literature review and analysis, drafting and critical revision and editing, and approval of the final version.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AUROC

area under the receiver operating characteristic curve

CI

confidence interval

DOR

diagnostic odds ratio

FN

false negative

FP

false positive

GC

gastric cancer

LR

likelihood ratio

QUADAS

quality assessment of diagnostic accuracy studies

SE

sensitivity

SP

specificity

TP

true positive

TN

true negative

TNM

tumor-node-metastasis

References

1 

Liu L, Wang S, Cao X and Liu J: Diagnostic value of circulating microRNAs for gastric cancer in Asian populations: A meta-analysis. Tumor Biol. 35:11995–2004. 2014. View Article : Google Scholar

2 

World Health Organization. Cancer Fact Sheets. http://globocan.iarc.fr/Pages/fact_sheets_population.aspx

3 

Duguan FU: Epigenetic alterations in gastric cancer (Review). Mol Med Rep. 12:3223–3230. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Wang QX, Zhu YQ, Zhang H and Xiao J: Altered MiRNA expression in gastric cancer: A systematic review and meta-analysis. Cell Physiol Biochem. 35:933–944. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Charalampakis N, Economopoulou P, Kotsantis I, Tolia M, Schizas D, Liakakos T, Elimova E, Ajani JA and Psyrri A: Medical management of gastric cancer: A 2017 update. Cancer Med. 7:123–133. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Hu N, Yin JF, Ji Z, Hong Y, Wu P, Bian B, Song Z, Li R, Liu Q and Wu F: Strengthening gastric cancer therapy by trastuzumab-conjugated nanoparticles with simultaneous encapsulation of anti-MiR-21 and 5-Fluorouridine. Cell Physiol Biochem. 44:2158–2173. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Huang S, Wang J, Li J, Luo Q, Zhao M, Zheng L, Dong X, Chen C, Che Y, Liu P, et al: Serum microRNA expression profile as a diagnostic panel for gastric cancer. Jpn J Clin Oncol. 46:8112016. View Article : Google Scholar : PubMed/NCBI

9 

Kitano S, Shiraishi N, Uyama I, Sugihara K and Tanigawa N: Japanese Laparoscopic Surgery Study Group: A multicenter study on oncologic outcome of laparoscopic gastrectomy for early cancer in Japan. Ann Surg. 245:68–72. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Uedo N, Takeuchi Y and Ishihara R: Endoscopic management of early gastric cancer: Endoscopic mucosal resection or endoscopic submucosal dissection: Data from a Japanese high-volume center and literature review. Ann Gastroenterol. 25:281–290. 2012.PubMed/NCBI

11 

Kaneko S and Yoshimura T: Time trend analysis of gastric cancer incidence in Japan by histological types. 1975–1989. Br J Cancer. 84:400–405. 2001. View Article : Google Scholar

12 

Bonenkamp JJ, Hermans J, Sasako M, van de Velde CJ, Welvaart K, Songun I, Meyer S, Plukker JT, Van Elk P, Obertop H, et al: Extended lymph-node dissection for gastric cancer. N Engl J Med. 340:908–914. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Garrido M, Bustos M, Orellana E, Madrid J, Galindo H, Sánchez C, Pimentel F, Guzmán S, Ibáñez L, Butte JM, et al: Postoperative radio chemotherapy in locally advanced gastric cancer. Rev Med Chil. 136:844–850. 2008.(In Spanish). PubMed/NCBI

14 

Madhavan D, Cuk K, Burwinkel B and Yang R: Cancer diagnosis and prognosis decoded by blood-based circulating microRNA signatures. Front Genet. 4:1162013. View Article : Google Scholar : PubMed/NCBI

15 

Komatsu S, Ichikawa D, Tsujiura M, Konishi H, Takeshita H, Nagata H, Kawaguchi T, Hirajima S, Arita T, Shiozaki A, et al: Prognostic impact of circulating miR-21 in the plasma of patients with gastric carcinoma. Anticancer Res. 33:271–276. 2013.PubMed/NCBI

16 

Chen CZ: MicroRNAs as oncogenes and tumor suppressors. N Engl J Med. 353:1768–1771. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993. View Article : Google Scholar : PubMed/NCBI

18 

Gorur A, Balci Fidanci S, Dogruer Unal N, Ayaz L, Akbayir S, Yildirim Yaroglu H, Dirlik M, Serin MS and Tamer L: Determination of plasma microRNA for early detection of gastric cancer. Mol Biol Rep. 40:2091–2096. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, et al: Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 105:10513–10518. 2008. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Deeks JJ and Morris JM: 6 Evaluating diagnostic tests. Baillière's Clin Obstet Gynaecol. 10:613–630. 1996. View Article : Google Scholar

22 

Deeks JJ: Systematic reviews in health care: Systematic reviews of evaluations of diagnostic and screening tests. BMJ. 323:157–162. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Jaeschke R, Guyatt GH and Sackett DL: Users' guides to the medical literature. III. How to use an article about a diagnostic test. B. What are the results and will they help me in caring for my patients? The Evidence-Based Medicine Working Group. JAMA. 271:703–707. 1994. View Article : Google Scholar : PubMed/NCBI

24 

Deeks JJ, Macaskill P and Irwig L: The performance of tests of publication bias and other sample size effects in systematic reviews of diagnostic test accuracy was assessed. J Clin Epidemiol. 58:882–893. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Higgins JP, Thompson SG, Deeks JJ and Altman DG: Measuring inconsistency in meta-analyses. BMJ. 327:557–560. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Jackson D, White IR and Thompson SG: Extending DerSimonian and Laird's methodology to perform multivariate random effects meta-analyses. Stat Med. 29:1282–1297. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Peng WZ, Ma R, Wang F, Yu J and Liu ZB: Role of miR-191/425 cluster in tumorigenesis and diagnosis of gastric cancer. Int J Mol Sci. 15:4031–4048. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Qiu X, Zhang J, Shi W, Liu S, Kang M, Chu H, Wu D, Tong N, Gong W, Tao G, et al: Circulating microRNA-26a in plasma and its potential diagnostic value in gastric cancer. PLoS One. 11:e01513452016. View Article : Google Scholar : PubMed/NCBI

29 

Tsujiura M, Komatsu S, Ichikawa D, Shiozaki A, Konishi H, Takeshita H, Moriumura R, Nagata H, Kawaguchi T, Hirajima S, et al: Circulating miR-18a in plasma contributes to cancer detection and monitoring in patients with gastric cancer. Gastric Cancer. 18:271–279. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Zhang J, Song Y, Zhang C, Zhi X, Fu H, Ma Y, Chen Y, Pan F, Wang K, Ni J, et al: Circulating MiR-16-5p and MiR-19b-3p as two novel potential biomarkers to indicate progression of gastric cancer. Theranostics. 5:733–745. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Tsai MM, Wang CS, Tsai CY, Huang CG, Lee KF, Huang HW, Lin YH, Chi HC, Kuo LM, Lu PH and Lin KH: Circulating microRNA-196a/b are novel biomarkers associated with metastatic gastric cancer. Eur J Cancer. 64:137–148. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Su ZX, Zhao J, Rong ZH, Wu YG, Geng WM and Qin CK: Diagnostic and prognostic value of circulating miR-18a in the plasma of patients with gastric cancer. Tumor Biol. 35:12119–12125. 2014. View Article : Google Scholar

33 

Valladares-Ayerbes M, Reboredo M, Medina-Villaamil V, Iglesias-Díaz P, Lorenzo-Patiño MJ, Haz M, Santamarina I, Blanco M, Fernández-Tajes J, Quindós M, et al: Circulating miR-200c as a diagnostic and prognostic biomarker for gastric cancer. J Transl Med. 10:1862012. View Article : Google Scholar : PubMed/NCBI

34 

Zhu X, Yang Z, Zhang J and Liu H: Investigation of microRNA-145 as a serum diagnostic and prognostic biomarker for gastric cancer: A Chinese cohort-based study. Int J Clin Exp Med. 10:9440–9447. 2017.

35 

Zhuang K, Han K, Tang H, Yin X, Zhang J, Zhang X and Zhang L: Up-regulation of plasma miR-23b is associated with poor prognosis of gastric cancer. Med Sci Monit. 22:356–361. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Hung PS, Chen CY, Chen WT, Kuo CY, Fang WL, Huang KH, Chiu PC and Lo SS: MiR-376c promotes carcinogenesis and serves as a plasma marker for gastric carcinoma. PLoS One. 12:e01773462017. View Article : Google Scholar : PubMed/NCBI

37 

Le Q, Jianhua N, Mulati, Yu X and Jiageng H: Increased miR-25 expression in serum of gastric cancer patients is correlated with CA19-9 and acts as a potential diagnostic biomarker. Open Medicine. 12:266–270. 2017. View Article : Google Scholar

38 

Li BS, Zhao YL, Guo G, Li W, Zhu ED, Luo X, Mao XH, Zou QM, Yu PW, Zuo QF, et al: Plasma microRNAs, miR-223, miR-21 and miR-218, as novel potential biomarkers for gastric cancer detection. PLoS One. 7:e416292012. View Article : Google Scholar : PubMed/NCBI

39 

Li F, Guo Y, Liu J and Zhang R: The significance of elevated plasma expression of microRNA 106b~25 clusters in gastric cancer. PLoS One. 12:e01784272017. View Article : Google Scholar : PubMed/NCBI

40 

Li M, Gu K, Liu W, Xie X and Huang X: MicroRNA-200c as a prognostic and sensitivity marker for platinum chemotherapy in advanced gastric cancer. Oncotarget. 8:51190–51199. 2017.PubMed/NCBI

41 

Hou X, Zhang M and Qiao H: Diagnostic significance of miR-106a in gastric cancer. Int J Clin Exp Pathol. 8:13096–13101. 2015.PubMed/NCBI

42 

Hou CG, Luo XY and Li G: Diagnostic and prognostic value of serum MicroRNA-206 in patients with gastric cancer. Cell Physiol Biochem. 39:1512–1520. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Zhang WH, Gui JH, Wang CZ, Chang Q, Xu SP, Cai CH, Li YN, Tian YP, Yan L and Wu B: The identification of miR-375 as a potential biomarker in distal gastric adenocarcinoma. Oncol Res. 20:139–147. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Song MY, Pan KF, Su HJ, Zhang L, Ma JL, Li JY, Yuasa Y, Kang D, Kim YS and You WC: Identification of serum microRNAs as novel non-invasive biomarkers for early detection of gastric cancer. PLoS One. 7:e336082012. View Article : Google Scholar : PubMed/NCBI

45 

Tsujiura M, Ichikawa D, Komatsu S, Shiozaki A, Takeshita H, Kosuga T, Konishi H, Morimura R, Deguchi K, Fujiwara H, et al: Circulating microRNAs in plasma of patients with gastric cancers. Br J Cancer. 102:1174–1179. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Wang B and Zhang Q: The expression and clinical significance of circulating microRNA-21 in serum of five solid tumors. J Cancer Res Clin Oncol. 138:1659–1666. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Zhu C, Ren C, Han J, Ding Y, Du J, Dai N, Dai J, Ma H, Hu Z, Shen H, et al: A five-microRNA panel in plasma was identified as potential biomarker for early detection of gastric cancer. Br J Cancer. 110:2291–2299. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Liu R, Zhang C, Hu Z, Li G, Wang C, Yang C, Huang D, Chen X, Zhang H, Zhuang R, et al: A five-microRNA signature identified from genome-wide serum microRNA expression profiling serves as a fingerprint for gastric cancer diagnosis. Eur J Cancer. 47:784–791. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Konishi H, Ichikawa D, Komatsu S, Shiozaki A, Tsujiura M, Takeshita H, Morimura R, Nagata H, Arita T, Kawaguchi T, et al: Detection of gastric cancer-associated microRNAs on microRNA microarray comparing pre- and post-operative plasma. Br J Cancer. 106:740–747. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Liu H, Zhu L, Liu B, Yang L, Meng X, Zhang W, Ma Y and Xiao H: Genome-wide microRNA profiles identify miR-378 as a serum biomarker for early detection of gastric cancer. Cancer Lett. 316:196–203. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Liu X, Kwong A, Sihoe A and Chu KM: Plasma miR-940 may serve as a novel biomarker for gastric cancer. Tumor Biol. 37:3589–3597. 2016. View Article : Google Scholar

52 

Hu Y, Wang J, Han Y, Liu Q and Niu Q: Serum miR-133a is down-regulated and associated with the diagnosis of patients with gastric cancer. Int J Clin Exp Pathol. 9:2015–2020. 2016.

53 

Sun Y, Ma J, Hu M, Zheng X, Li J and Gu Wei: Level of miR-183 in peripheral blood of the patients with gastric cancer. J Shanghai Jiaotong Univ. 37:75–79. 2017.

54 

Li B and Zhang H: Plasma microRNA-320 is a potential diagnostic and prognostic bio-marker in gastric cancer. Int J Clin Exp Pathol. 10:7356–7361. 2017.

55 

Wu J, Li G, Yao Y, Wang Z, Sun W and Wang J: MicroRNA-421 is a new potential diagnosis biomarker with higher sensitivity and specificity than carcinoembryonic antigen and cancer antigen 125 in gastric cancer. Biomarkers. 20:58–63. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Zeng Q, Jin C, Chen W, Xia F, Wang Q, Fan F, Du J, Guo Y, Lin C, Yang K, et al: Downregulation of serum miR-17 and miR-106b levels in gastric cancer and benign gastric diseases. Chin J Cancer Res. 26:711–716. 2014.PubMed/NCBI

57 

Zhou H, Xiao B, Zhou F, Deng H, Zhang X, Lou Y, Gong Z, Du C and Guo J: MiR-421 is a functional marker of circulating tumor cells in gastric cancer patients. Biomarkers. 17:104–110. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Zhou X, Zhu W, Li H, Wen W, Cheng W, Wang F, Wu Y, Qi L, Fan Y, Chen Y, et al: Diagnostic value of a plasma microRNA signature in gastric cancer: A microRNA expression analysis. Sci Rep. 5:112512015. View Article : Google Scholar : PubMed/NCBI

59 

Wu J, Li G, Wang Z, Yao Y, Chen R, Pu X and Wang J: Circulating MicroRNA-21 is a potential diagnostic biomarker in gastric cancer. Dis Markers. 2015:4356562015. View Article : Google Scholar : PubMed/NCBI

60 

Wu D, Cao G, Huang Z, Jin K, Hu H, Yu J and Zeng Y: Decreased miR-503 expression in gastric cancer is inversely correlated with serum carcinoembryonic antigen and acts as a potential prognostic and diagnostic biomarker. OncoTargets Ther. 10:129–135. 2017. View Article : Google Scholar

61 

Wang H, Wang L, Wu Z, Sun R, Jin H, Ma J, Liu L, Ling R, Yi J, Wang L, et al: Three dysregulated microRNAs in serum as novel biomarkers for gastric cancer screening. Med Oncol. 31:2982014. View Article : Google Scholar : PubMed/NCBI

62 

Shin VY, Ng EK, Chan VW, Kwong A and Chu KM: A three-miRNA signature as promising non-invasive diagnostic marker for gastric cancer. Mol Cancer. 14:2022015. View Article : Google Scholar : PubMed/NCBI

63 

Cui YJ, Xie XH, Xing YF, Yuan ZH, Wu QY and Wei YM: Feasibility study on plasmatic microRNA-27b-3p as a potential biomarker for early diagnosis of gastric cancer. Tumor. 35:183–189. 2015.

64 

Fu Z, Qian F, Yang X, Jiang H, Chen Y and Liu S: Circulating miR-222 in plasma and its potential diagnostic and prognostic value in gastric cancer. Med Oncol. 31:1642014. View Article : Google Scholar : PubMed/NCBI

65 

Liu H, Yang Z, Zhang J and Zhu X: MicroRNA-217 in plasma: A potential biomarker in gastric cancer. Int J Clin Exp Med. 10:3313–3320. 2017.

66 

Zhou H, Guo JM, Lou YR, Zhang XJ, Zhong FD, Jiang Z, Cheng J and Xiao BX: Detection of circulating tumor cells in peripheral blood from patients with gastric cancer using microRNA as a marker. J Mol Med (Berl). 88:709–717. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Zhou X and Zhang G: Exosome-mediated transfer of miR-223 increases tumor malignancy in gastric cancer. J Gastroen Hepatol (Australia). 31:45–46. 2016.

68 

Zheng Y, Cui L, Sun W, Zhou H, Yuan X, Huo M, Chen J, Lou Y and Guo J: MicroRNA-21 is a new marker of circulating tumor cells in gastric cancer patients. Cancer Biomark. 10:71–77. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Li H, Wu Q, Li T, Liu C, Xue L, Ding J, Shi Y and Fan D: The miR-17-92 cluster as a potential biomarker for the early diagnosis of gastric cancer: Evidence and literature review. Oncotarget. 8:45060–45071. 2017.PubMed/NCBI

70 

Park JL, Kim M, Song KS, Kim SY and Kim YS: Cell-Free miR-27a, a potential diagnostic and prognostic biomarker for gastric cancer. Genomics Inform. 13:70–75. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Li C, Li JF, Cai Q, Qiu QQ, Yan M, Liu BY and Zhu ZG: MiRNA-199a-3p: A potential circulating diagnostic biomarker for early gastric cancer. J Surg Oncol. 108:89–92. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Li C, Li JF, Cai Q, Qiu QQ, Yan M, Liu BY and Zhu ZG: MiRNA-199a-3p in plasma as a potential diagnostic biomarker for gastric cancer. Ann Surg Oncol. 20((Suppl 3)): S397–S405. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Liu S, Suo J, Wang C, Sun X, Wang D, He L, Zhang Y and Li W: Prognostic significance of low miR-144 expression in gastric cancer. Cancer Biomark. 20:547–552. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Lurje G, Schiesser M, Claudius A and Schneider PM: Circulating tumor cells in gastrointestinal malignancies: Current techniques and clinical implications. J Oncol. 2010:3926522010. View Article : Google Scholar : PubMed/NCBI

76 

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

77 

Redova M, Sana J and Slaby O: Circulating miRNAs as new blood-based biomarkers for solid cancers. Future Oncol. 9:387–402. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Shen J, Stass SA and Jiang F: MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett. 329:125–136. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J, Poon TC, Ng SS and Sung JJ: Differential expression of microRNAs in plasma of patients with colorectal cancer: A potential marker for colorectal cancer screening. Gut. 58:1375–1381. 2009. View Article : Google Scholar : PubMed/NCBI

80 

Zanutto S, Pizzamiglio S, Ghilotti M, Bertan C, Ravagnani F, Perrone F, Leo E, Pilotti S, Verderio P, Gariboldi M and Pierotti MA: Circulating miR-378 in plasma: A reliable haemolysis independent biomarker for colorectal cancer. Br J Cancer. 110:1001–1007. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

82 

Matsushima K, Isomoto H, Inoue N, Nakayama T, Hayashi T, Nakayama M, Nakao K, Hirayama T and Kohno S: MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int J Cancer. 128:361–370. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 41 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei H, Pu K, Liu XG, Li BX, Zhang HS, Wang H, Wang H, Sun WM and Wang YP: The diagnostic value of circulating microRNAs as a biomarker for gastric cancer: A meta‑analysis. Oncol Rep 41: 87-102, 2019
APA
Wei, H., Pu, K., Liu, X., Li, B., Zhang, H., Wang, H. ... Wang, Y. (2019). The diagnostic value of circulating microRNAs as a biomarker for gastric cancer: A meta‑analysis. Oncology Reports, 41, 87-102. https://doi.org/10.3892/or.2018.6782
MLA
Wei, H., Pu, K., Liu, X., Li, B., Zhang, H., Wang, H., Wang, H., Sun, W., Wang, Y."The diagnostic value of circulating microRNAs as a biomarker for gastric cancer: A meta‑analysis". Oncology Reports 41.1 (2019): 87-102.
Chicago
Wei, H., Pu, K., Liu, X., Li, B., Zhang, H., Wang, H., Wang, H., Sun, W., Wang, Y."The diagnostic value of circulating microRNAs as a biomarker for gastric cancer: A meta‑analysis". Oncology Reports 41, no. 1 (2019): 87-102. https://doi.org/10.3892/or.2018.6782