Next Article in Journal
Comprehensive Perspective for Lung Cancer Characterisation Based on AI Solutions Using CT Images
Previous Article in Journal
Autologous Fat Transplantation for Thumb Carpometacarpal Joint Osteoarthritis (Liparthroplasty): A Case Series with Two Years of Follow-UP
Previous Article in Special Issue
Belching in Gastroesophageal Reflux Disease: Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular miRNAs for the Management of Barrett’s Esophagus and Esophageal Adenocarcinoma: A Systematic Review

by
Kazumi Inokuchi
1,
Takahiro Ochiya
2 and
Juntaro Matsuzaki
1,2,*
1
Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
2
Department of Molecular and Cellular Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(1), 117; https://doi.org/10.3390/jcm10010117
Submission received: 3 October 2020 / Revised: 26 December 2020 / Accepted: 28 December 2020 / Published: 31 December 2020
(This article belongs to the Special Issue New Advances in Gastroesophageal Reflux Disease)

Abstract

:
Esophageal adenocarcinoma (EAC), the major histologic type of esophageal cancer (EC) in Western countries, is a disease with a poor prognosis, primarily due to usual diagnosis at an advanced stage. The prevalence of EAC has increased in recent years, both in Western countries and in Asia. Barrett’s esophagus (BE) is a precursor lesion of EAC. Therefore, early detection and proper management of BE and EAC is important to improve prognosis. Here, we systematically summarize current knowledge about the potential utility of extracellular microRNAs (miRNAs), which are thought to be non-invasive biomarkers for many diseases, for these purposes. A search of the PubMed and Embase databases identified 22 papers about extracellular miRNAs that have potential utility for management of EAC. Among them, 19 were EAC-related and ten were BE-related; some of these dealt with both conditions. The articles included studies reporting diagnosis, prognosis, and treatment responses. Multiple papers report dysregulation of miR-194-5p in BE and miR-21-5p, -25-3p, and -93-5p in EAC. Although it will take time to utilize these miRNAs in clinical practice, they are likely to be useful non-invasive markers in the future.

1. Introduction

Esophageal adenocarcinoma (EAC) is a major histologic type of esophageal cancer (EC), the incidence of which varies geographically according to race and gender [1]. The rate of EAC in Western Europe, Australia, and North America is increasing rapidly [2,3,4]. By 2030, 1 in 100 men in the Netherlands and United States are predicted to be diagnosed with EAC during their lifetime [5]. In Japan, although EAC accounts for less than 10% of EC cases, the incidence is increasing [6]. The increase can be explained by decreased prevalence of Helicobacter pylori (H. pylori) infection, although the relationship between H. pylori and EAC occurrence remains controversial. [7,8,9].
The prognosis of EAC is poor if diagnosed at the advanced stage, by which time metastasis or invasion has occurred. The 5-year survival rate for EAC patients is 80.5% at stage I, and 45.1%, 17.6%, and 2.1% at stages II, III, and IV, respectively [3]. However, increased rates of early detection in European countries, the United States, and China have improved the 5-year survival rate from less than 5% in the 1960s to about 20% in 2000 [10,11,12]. Thus, early identification of EAC and identification of high-risk groups are important.
Barrett’s esophagus (BE) denotes metaplasia from squamous epithelium into columnar epithelium; BE is classified into short-segment BE (SSBE) and long-segment BE (LSBE). BE is considered to be a precancerous lesion of EAC. Both SSBE and LSBE carry a risk of carcinogenesis [13], although the risk is lower for SSBE [14]. BE and EAC are induced by reflux of both gastric and bile acids [15], irrespective of the severity of reflux symptoms and esophagitis [4,16]. Similar to the adenoma-carcinoma sequence of colorectal cancer, EAC develops through metaplasia, low-grade dysplasia (LGD), high-grade dysplasia (HGD), intraepithelial adenocarcinoma, and finally invasive adenocarcinoma. Jason et al. reported that BE develops in 6–14% of patients with GERD, and that 0.5–1% of these will develop adenocarcinoma [4,17]. Thus, to facilitate early detection of EAC, it is important to identify patients with BE who are at high risk of EAC.
Esophagogastroduodenoscopy is the standard method for early detection of BE and EAC; no non-invasive and reliable screening method has been established to date. Although various efforts and trials, such as Seattle protocol, web-based teaching tools [18], and computer-aided detection [19], have been made to improve detection of BE and EAC, the examination itself is invasive and relies heavily on the skill of the operator. The Cytosponge, a gelatin-coated sponge attached to a string that collects cytologic specimens from the esophageal mucosa when withdrawn, may have the potential to replace traditional endoscopic screening and be more cost-effective [20]. However, it is not used widely.
Recently, circulating cell-free DNA and RNA have attracted attention as new biomarkers. MiRNA is a single-stranded non-coding RNA comprising 17–25 bases, which regulates protein translation. It is suggested that tumor tissues may secrete aberrant miRNAs to create a microenvironment conducive to growth. Furthermore, miRNAs are actively secreted into blood, urine, and saliva by extracellular vesicles. Thus, miRNAs may be novel biomarkers for various pathological conditions, including not only cancer but also inflammatory and infectious diseases [21]. Since the discovery of extracellular miRNAs, many studies have actively investigated BE and EAC-specific miRNAs for diagnostic purposes. Here, we systematically reviewed the literature regarding extracellular miRNAs related to BE and EAC.

2. Experimental Section

Methods

The PubMed and Embase databases were searched using the terms “(esophagus OR esophageal) AND (Barrett’s OR adenocarcinoma) AND (microRNA OR miRNA) AND (circulating OR serum OR plasma OR urine)”. Of these, duplications, review articles, studies of intracellular miRNAs only, and studies not investigating BE or EAC (i.e., investigations of esophageal squamous cell carcinoma only) were excluded (Figure 1). The contents of these papers were organized and are described below.

3. Results

We identified 35 reports in PubMed and 78 reports in Embase. After applying the exclusion criteria, we identified 22 papers (including abstracts for conference presentations). We classified these into those related to diagnosis of BE, those related to diagnosis of EAC, those related to therapeutic effects, and those related to prognosis of EAC (Figure 2).

3.1. Detection of BE

Six studies reported diagnosis of GERD and BE; some of these also mentioned EAC. Those comparing EAC and BE are described in the next section. Here, we describe studies that investigated miRNAs that distinguish individuals with GERD or BE from normal healthy subjects (Table 1).
In 2013, Bansal et al. [22] compared serum levels of miRNAs in 20 BE patients and 19 patients with GERD (grade B or higher). Compared with the GERD group, the BE group had significantly higher levels of miR-15a-5p (sensitivity, 75%; specificity, 75%; area under the receiver operating characteristic curve (AUC), 0.68 (0.60–0.86, p = 0.01)) and miR-196a-5p (sensitivity, 70%; specificity, 74%; AUC, 0.72 (0.61–0.89, p = 0.03)). A combination of miR-15a and miR-196a can be used to distinguish BE patients from GERD patients, with a sensitivity of 75%, a specificity of 85%, and an AUC of 0.79 (0.64–0.95, p = 0.003).
In 2016, Bus et al. [23] identified four BE-specific miRNAs by comparing plasma miRNAs from BE and EAC with those from healthy controls. Further validation in 115 patients and controls showed that miRNA-194-5p and miRNA-451a were up-regulated, and miRNA-136-5p was down-regulated, in BE compared with controls. Furthermore, a combination of three or more miRNAs showed good diagnostic performance, with an AUC of 0.832.
In the same year, Cabbi et al. [24] compared miRNA expression in serum and esophageal mucosal biopsy tissues from patients with BE, columnar-lined esophagus (CLE), and esophagitis. Similar to the results from tissues, circulating levels of miR-143-3p, -194-5p, and -215-5p were up-regulated in the serum of patients with BE, and were higher than those in patients with CLE or esophagitis. MiR-194-5p and miR-215-5p were also significantly up-regulated in patients with CLE compared with those with esophagitis, suggesting that CLE is an intermediate condition between BE and esophagitis.
In 2017, Fassan et al. [25] investigated expression of plasma miR-223-3p in BE and EAC, and found that expression of miR-223-3p was significantly associated with the degree of intestinal metaplasia in those with atrophic gastritis and LSBE.
Pavlov et al. [26] compared serum miRNAs from patients with normal squamous epithelium (SE), BE, HGD, and EAC. MiR-320e was significantly down-regulated in the BE group compared with the SE (p ≤ 0.001; AUC, 0.790) and HGD (p ≤ 0.005; AUC, 0.786) groups, and miR-199a-3p was significantly down-regulated compared with the SE group (p ≤ 0.001; AUC, 0.813).
In 2019, Wang et al. [27] found higher levels of serum miR130a-3p in patients with low-grade and high-grade dysplastic BE than in serum from patients with metaplastic epithelium.
Table 1. MiRNAs used to detect BE.
Table 1. MiRNAs used to detect BE.
Author, Year [Reference]Sample SizeSampleUp-RegulatedDown-Regulated
Bansal et al.
2013 [22]
BE (n = 20)
GERD (n = 19)
Serum-15a-5p
-195a-5p
Bus et al.
2014 [28]
BE (n = 8)
Healthy (n = 6)
Plasma-194-5p, -451a-136-5p
Cabbi et al.
2016 [24]
BE (n = 8)
CLE (n = 12)
Esophagitis (n = 10)
Plasma-143-3p
-215-5p
-194-5p
Pavlov et al.
2018 [26]
BE (n = 27)
SE (n = 19)
HGD (n = 17)
Serum -320e
-199a-3p
Wang et al. 2019 [27]BE (n = 6)
Healthy (n = 6)
Serum-130a-3p
BE, Barrett’s esophagus; CLE, columnar-lined esophagus; SE, squamous epithelium; HGD, high-grade dysplasia.

3.2. Detection of EAC

There were 14 publications about the utility of miRNAs for diagnosis of EAC. The controls were mostly healthy individuals or BE patients, and one report used saliva samples (Table 2).
In 2011, Song et al. [29] extracted total RNA from serum samples obtained from EAC patients and healthy controls, and from cancer tissues and normal tissues of EAC patients. Some miRNAs showed significant differences in serum levels between the EAC and healthy groups (AUC, 0.885; sensitivity, 92%; specificity, 83%). They reported that measurement of serum miRNAs is likely to lead to early diagnosis of EAC and to an improved prognosis (the specific name of miRNA was not reported).
In 2013, Xie et al. [30] analyzed saliva miRNAs from EC patients and healthy subjects, and verified their presence in both whole saliva and saliva supernatant samples from 39 EC patients and 39 healthy controls. MiR-10b-3p, -144-3p, and -451a in whole saliva, and miR-10b-3p, -144-3p, -21-5p, and -451a in saliva supernatant, were significantly up-regulated in EC patients (sensitivity, 89.7%, 92.3%, 84.6%, 79.5%, 43.6%, 89.7%, and 51.3%, respectively, and specificity, 57.9%, 47.4%, 57.9%, 57.9%, 89.5%, 47.4%, and 84.2%, respectively). Although this report also included SCC, the authors reported no differences in expression of miRNA between EAC and SCC.
In 2015, Chiam et al. [31] verified the presence of serum exosome miRNAs in 18 cases of locally advanced EAC, ten cases of BE, and 19 healthy controls, and identified a multi-biomarker panel (RNU6-1/miR-16-5p, miR-25-3p/miR-320a, let-7e-5p/miR-15b-5p, miR-30a-5p/miR-324-5p, and miR-17-5p/miR-194-5p) that is more specific than any single miRNA. They reported that this panel distinguished EAC patients from healthy controls and BE patients with high specificity and sensitivity (ROC = 0.99; 95% confidence interval (CI), 0.96–1.0).
Warnecke-Eberz et al. [32] reported overexpression of serum exosome-derived miR-223-5p and miR-483-5p in EAC patients. Furthermore, regarding differences in TNM classification, miR-223-5p expression was higher in T2 (p = 0.006) than in T3. These data suggest that miR-223 is secreted at high levels during the early stages of tumor development.
Bus et al. [23] compared BE, EAC, and control plasma miRNAs, and reported that miR-382-5p was significantly up-regulated, and miR-133a-3p was down-regulated, in EAC patients, and that combinations of three or more miRNAs could distinguish EAC from controls (AUC, 0.846) and BE from EAC (AUC, 0.797).
In 2016, Yan et al. [33] reported that exosomes extracted from EAC-conditioned medium or serum from EAC patients contained significantly higher levels of miR-93-5p and miR-21-5p than normal medium or normal serum. In addition, they cultivated normal enteroids and BE organoids for 3 weeks in EAC-conditioned medium. They found that EAC-conditioned medium induced dysplasia and proliferation of enteroids and BE organoids. Therefore, they hypothesized that exosomes from cancer cells play a role in induction of morphological changes in organoids and promote their growth.
Zhang et al. [34] compared circulating miRNAs in serum from EAC patients and healthy controls, and found that miR-25-3p and miR-151a-3p were significantly up-regulated, and that miR-100-5p and miR-375-3p were significantly down-regulated in EAC patients.
Chen et al. [35] isolated exosomes from serum samples from EAC patients and controls, and reported that miR-21-5p, -16-5p, -25-3p, and -155-5p were significantly up-regulated, and miR-192-5p was significantly down-regulated, in EAC patients.
We [36] also analyzed tissue and blood miRNAs from human interleukin-1β (IL-1β) transgenic mice (L2-IL-1β mice) as a model of BE. We then compared expression of human plasma miRNAs in eight EAC patients, eight BE patients, and six healthy individuals using a public dataset. Compared with the BE group, levels of miR-128-3p and miR-328-3p were higher in the EAC group, and those of miR-143-3p, -144-3p, -15a-5p, -1-3p, and -133b were lower.
In 2018, Miyoshi et al. [37] profiled miRNAs expressed in 42 pairs of EAC and normal tissues, and validated them in 44 EAC, 20 BE, and 30 control sera. Six miRNAs (miR-106b-5p, -146a-5p, -15a-5p, -18a-5p, -21-5p, and -93-5p) were significantly up-regulated and could be used to distinguish EAC patients from healthy subjects (AUC, 0.86).
In 2019, Wang et al. [27] profiled BE and healthy subjects, and compared serum miRNAs among EAC, BE, and healthy groups. They found that miR130a-3p was significantly up-regulated in the EAC group, and that expression of miR130a-3p increased gradually from early (I, II) to advanced (III, IV) stage. As we mentioned in the BE section, they also showed that levels of miR130a-3p were significantly higher in patients with LGD and HGD BE than in those with metaplasia (sensitivity, 70.5%; specificity, 62.5%), suggesting that increased miR130a-3p expression in plasma may correlate with early EAC.
Craig et al. [38] compared miRNAs in serum from five controls, nine GERD, seven BE, five BE with LGD, and five EAC patients with those in biopsy tissues. Some characteristic miRNAs were identified in biopsy tissues, but there was no difference between expression of serum miRNAs between EAC patients and controls.
Fassan et al. [39] also compared serum miRNA from patients with BE, HG-IEN (dysplastic or intraepithelial neoplasia), and EAC. Seven (miR-92a-3p, -151a-5p, -362-3p, -345-3p, -619- 3p, -1260b, and -1276) were up-regulated in the HG-IEN/EAC samples compared with the non-dysplastic BE samples, whereas three (miR-381-3p, -502-3p, and -3615) were down-regulated.
Table 2. MiRNAs used to detect EAC.
Table 2. MiRNAs used to detect EAC.
Author, Year [Reference]Sample SizeSampleUp-RegulatedDown-Regulated
Xie et al.
2013 [30]
SCC (n = 32)
EAC (n = 4)
Healthy (n = 19)
Whole
saliva
-10b-3p, -144-3p,
-451a
Saliva supernatant-10b-3p, -144-3p,
-21-5p, -451a
Chiam et al.
2015 [31]
EAC (n = 18)
BE (n = 10)
Healthy (n = 19)
SerumRNU6-1/-16-5p,
-25-3p/-320a
let-7e-5p/-16b-5p,
-30a-5p/-324-5p,
-17-5p/-194-5p
Warnecke-Eberz et al.
2015 [32]
EAC (n = 59)
Healthy (n = 4
Serum-223-5p, -483-5p
Bus et al.
2016 [23]
EAC (n = 59)
BE (n = 41)
Healthy (n = 15)
Plasma-382-5p-133a-3p
Yan et al.
2016 [33]
Medium
serum
-93-5p, -21-5p
Zhang et al.
2016 [34]
EAC (n = 10)
Healthy (n = 11)
Serum-25-3p, 151a-3p-100-5p, -375-3p
Chen et al.
2017 [35]
EAC (n = 9)
Healthy (n = 9)
Serum-21-5p, -16-5p,
-25-3p, -155-5p
-192-5p
Fassan et al.
2017 [25]
EAC (n = 30)
Dyspeptic patients (n = 20)
Atrophic gastritis (n = 15)
BE (n = 15)
Plasma-223-3p
Matsuzaki et al.
2017 [36]
EAC (n = 8)
BE (n = 8)
Healthy (n = 6)
Plasma-128-3p, -328-3p-143-3p, -144-3p,
-15a-5p, -1-3p,
-133b
Miyoshi et al.
2018 [37]
EAC (n = 44)
BE (n = 20)
Healthy (n = 30)
Serum,-106b-5p, -93-5p,
-146a-5p, -15a-5p,
-18a-5p, -21-5p,
Wang et al.
2019 [27]
EAC (n = 40)
BE (n = 60)
Healthy (n = 30)
Serum-130a-3p
Fassan et al.
2020 [39]
EAC (n = 8)
BE (n = 12)
HG-IEN (n = 5)
Serum-92a-3p, -151a-5p,
-362-3p, -345-3p,
-619-3p, 1260b,
-1276
-381-3p, -502-3p,
-3615
EAC, esophageal adenocarcinoma; BE, Barrett’s esophagus; HGD, high-grade dysplasia.

3.3. MiRNAs Related to Prognosis and Treatment Responses in EAC Patients

There have been some attempts to use miRNAs not only for diagnosis of EAC but also for treatment responses and prognosis. With respect to recurrence, Gu et al. [40] investigated the role of serum miRNAs as predictors of recurrence. They profiled serum miRNAs from 72 EAC patients (32 with recurrence and 40 without) and validated them in 329 EAC patients (132 with recurrence and 197 without). The results showed that serum miR-331-3p was significantly down-regulated in patients with recurrence, making it a useful biomarker for identifying EAC patients at high risk of recurrence. The following four studies examined treatment response and prognosis.
Odenthal et al. [41] examined serum miRNAs in 50 patients with locally advanced EAC who underwent neoadjuvant chemotherapy and surgery, and assessed whether serum miRNAs were useful indicators of treatment response. No serum miRNAs were associated with a therapeutic response, but there was a significant correlation between expression of miR-192-5p and miR-222-3p and T classification, and between expression of miR-302c-3p and miR-222-3p and overall survival (OS). They reported that low miR-222-3p expression and high 302c-3p expression may suggest significantly better OS.
In 2014, Pu et al. [42] attempted to identify serum miRNAs as diagnostic, prognostic, and predictive biomarkers for EAC. When comparing miRNAs in the EAC and the control groups, they identified four (miR-126-3p, -142-3p, -331-3p, and -18a-5p) that were specific for EAC (p < 0.05). In addition, five miRNAs were significantly associated with survival, two of which were associated with recurrence (p < 0.05) (the specific miRNAs were not identified). Also, in cases with high miR-30c-5p expression, the risk of death (HR = 0.28, 95% CI = 0.12–0.65, p = 0.003) and recurrence (HR = 0.49; 95% CI = 0.24–0.99, p = 0.047) was lower than in cases with low expression (“risk of death” was not defined). Among 118 patients who received chemo/chemoradiation therapy (93% and 84% received 5FU and platinum, respectively), those in both treatment groups with high expression of miR-30c-5p had a reduced risk of mortality. In the platinum group, miR-26a-5p was significantly related to increased risk of death (HR = 5.78; 95% CI = 1.59–21.06, p = 0.008) and recurrence (HR = 7.76; 95% CI = 2.30–26.18, p = 0.001), and miR-142-3p was associated with an increased risk of recurrence (HR = 2.67; 95% CI = 1.09–6.56, p = 0.032). By contrast, in the 5FU group, miR-127-3p and miR-486-5p were significantly associated with both survival and recurrence (it is unclear whether high or low expression was important).
In 2015, Zhai et al. [43] investigated whether serum miRNA profiles could predict the prognosis of EAC together with H. pylori infection status. Serum samples from EAC patients and healthy individuals were used to profile miRNAs, and serum from other EAC patients were evaluated to examine the association between survival and the presence or absence of H. pylori infection (H. pylori was detected by immunoblotting). Overall, serum miRNAs were not significantly associated with the prognosis of EAC in the H. pylori-positive group. However, 16 miRNAs (miR-1253, -1273d, -187-5p, -1912, -200b-5p, -2276, -3147, -324-3p, -326, -3652, -3935, -4267, -4274, -4286, -4323, and -640) were significantly associated with OS (all p < 0.05) in the H. pylori-negative group. This suggests that miRNAs affect the survival rate of those with EAC and may be related to the presence or absence of H. pylori infection.
Petrick et al. [44] used US multicenter population-based serum samples from patients enrolled between 1993 and 1995 to compare miRNAs in EAC and healthy individuals, and to investigate survival rates. In the EAC group, 68 serum miRNAs were significantly up-regulated and 11 serum miRNAs were down-regulated. MiR-4253 and miR-1238-5p were associated with survival after EAC diagnosis (HR = 4.85, 95% CI, 2.30–10.23, BH-adjusted p = 0.04; and HR = 3.81, 95% CI, 2.02–7.19, BH-adjusted p = 0.04, respectively). This study suggests that circulating miRNAs may be associated with survival after EAC diagnosis.

4. Discussion

4.1. BE diagnosis

Seven miRNAs (miR-15a-5p, -130a-3p, -143-3p, -194-5p, 195a-5p, -215-5p, and -451a) were up-regulated and three miRNAs (miR-136-5p, -320e, and -199a-3p) were down-regulated. MiR-15a-5p [37], -130-3p [27], and -451a [30] are also up-regulated in EAC, whereas miR-143 is down-regulated.
MiR-15a-5p is involved in colon cancer, cervical cancer, and cervical cell cancer [45,46,47,48], and the main function of miR-130a-3p is to inhibit tumor progression [49,50]. MiR-143-3p [51,52,53] and miR-451a are tumor suppressors that correlate with non-small cell lung cancer (NSCLC) and colorectal cancer [53,54]. MiR-194-5p inhibits cell migration and invasion during gastric cancer progression by down-regulating FoxM1 [55]. MiR-136-5p suppresses lung adenocarcinoma [56] and renal cell cancer [57]. MiR-199a-3p suppresses EAC [58] and other cancers such as gastric cancer [59]. A report suggests that tissue miR-215-5p is specific for BE [60]; however, others suggest that it suppresses some tumors [61,62]. Previous studies show that expression of miR-320e is higher in EAC tissues than in BE tissues; high miR-320e tissue levels are associated with an adverse prognosis in those with colon cancer [63], although the underlying mechanism is unclear (Table 3).
Based on the above, levels of miRNA in blood differ between BE and healthy subjects; two papers report upregulation of miR-194-5p in BE [23,24]. MiR-194-5p and miR-215-5p levels are higher in those with BE, and even in those with CLE, than in controls [24]. It is thought that specific serum miRNAs start to rise at the very early stage, when the SE begins to show morphological changes, and that this may be useful for diagnosis of early dysplasia and BE.

4.2. EAC Diagnosis

Many miRNAs are associated with EAC diagnosis, among which miR-21-5p, -25-3p, and -93-5p are mentioned in more than one paper. MiR-21-5p was reported in four papers [30,33,35,37]; indeed, it is one of the most commonly observed aberrant miRNAs in human cancer. Numerous studies show that miR-21-5p promotes carcinogenesis and tumor progression by targeting genes encoding PDCD4, PTEN, BTG2, FasL, IGFBP3, TGF-β1, FBXO11, and TIMP3 [66,67,68,69,70,71]. Many studies explored the function of miR-21-5p with respect to prognosis and diagnosis. With respect to EAC, miR-21-5p upregulation promotes metastasis of EAC by regulating cancer cell apoptosis [72]; it is also an independent prognostic biomarker for disease-specific survival [73]. More than two papers report upregulation of miR-25-3p [31,34] and miR-93-5p [33,37]. High expression of miR-25-3p in EAC tissues is reported in several papers; this miRNA promotes cell migration and invasion of SCC by targeting CDH1 [74]. Mir-93-5p was identified by studies examining EAC tissue miRNA; the combination of miR-25/miR-93 establishes an immunosuppressive microenvironment in breast cancer by inactivating the cGAS pathway [75], and it also reported to have the promotion function on cervical cancer by targeting THB2/Matrix metalloproteinases (MMPS) pathway [76] MiR-15a-5p is both up-regulated [37] and down-regulated [36,77] in cancer serum; some reports suggest that it acts as a tumor suppressor [47,78], whereas some suggest that it acts as a tumor promoter [48]. Further research is necessary. MiR-223-5p is expressed at higher levels at T2 than at T3. MiR-223-5p inhibits tumor progression [79], whereas miR-130a-3p expression increases as the tumor stage progresses [27], suggesting that it promotes tumor growth [64] (Table 4).
Many studies show that extracellular miRNA levels are significantly different between EAC, BE, and controls, but there is also a report that the utility of serum miRNAs is limited [38]. The roles of miR-21-5p, -25-3p, and -93-5p in cancer have been reported in multiple papers [76]. In addition, serum miR-223-5p levels increase more quickly at the early stage of EAC [32], and serum levels of miR-130a increase as tumor stage progresses [27]. Thus, miRNAs are useful markers for early detection and staging of EAC.

4.3. EAC Recurrence

Two papers reported that serum levels of miR-331-3p, -30c-5p, 142-3p, 127-3p, 486-5p, and -26a-5p correlate with recurrence of EAC [40,42]. There are few reports of cellular or extracellular miRNAs in the context of EAC recurrence. Matsui et al. [80] identified miR-652-5p, -7-2-3p, -3925-3p, and -219-3p as being prognostic for recurrence in patients with stage II/III EAC. But in the two papers we found here, the same extracellular miRNAs were not reported.
MiR-142-3p suppresses growth of colorectal cancer [81] and breast cancer [82] cells by targeting CDK4, Beach-1, and CDC25C [83]. MiR-331-3p is useful for diagnosis of EAC [42] and is known to promote tumor progression [83]. As we mentioned in the previous paragraph about EAC recurrence, four (miR-30c-5p, -127-3p, -486-5p, and -26a-5p) serum miRNAs are associated with OS/prognosis (Table 5).

4.4. Survival and Prognosis

Among the miRNAs associated with OS or prognosis, miR-30c-5p, -26a-5p, -127-3p, and -486-5p are also associated with recurrence. Serum miR-222-3p is also associated with T classification [41,42].
MiR-30c-5p suppresses tumorigenesis in gastric cancer [89] and renal cell carcinoma (RCC) [90]. MiR-331-3p regulates cancer growth by targeting HER2, overexpression of which promotes cancer cell proliferation, resistance to apoptosis, angiogenesis, and metastasis [95]. MiR-331-3p expression is down-regulated in various cancers (e.g., cervical cancer [84], colorectal cancer [85], ovarian cancer [86], gastric cancer [87], prostate cancer, and glioblastoma). MiR-26a-5p inhibits migration and invasion of cancer cells by targeting the TMEM184B gene [88]. MiR-127-3p acts as a tumor growth suppressor by targeting KIF3, ITGA6, and BAG5 in SCC [96], osteosarcoma [92], and ovarian cancer [93]. MiR-486-5p stimulates cell proliferation, migration, and invasion by inhibiting PTEN expression and by activating the oncogenic PI3K/AKT pathway [97,98], although one study concluded that it works to suppress tumors [99]; further verification is required (Table 5).
In addition, Zhai et al. reported that miRNAs that predict prognosis differed according to H. pylori infection status [43]. H. pylori infection induces chronic gastritis and metaplasia, which can alter miRNA expression profiles in human gastric mucosa [100,101]. Wang et al. reported that miRNA expression in 53 gastric cancer tissues differed between H. pylori-positive and H. pylori-negative patients [102]. Importantly, most patients with EAC do not have H. pylori infection because H. pylori infection reduces gastric acid secretion and acid regurgitation into the esophagus. [103]. Differences in expression of miRNAs associated with the prognosis of EAC might be due to different etiologies of EAC.
From the above, we can see that many miRNAs are associated with tumor progression. Thus, increased or decreased levels of these in the blood may reflect the growth rate of the tumor, and will therefore reflect survival or recurrence rates.

4.5. Potential of Extracellular miRNAs as BE/EAC Diagnosis/Prediction and Prognosis Markers

Detection of EAC by endoscopy requires skill and continuous training. In Western countries, although several screening methods for EAC have been suggested, the most cost-effective strategies for risk stratification and surveillance intervals remain controversial [17]. In fact, the detection rate of EAC by surveillance alone is low; less than 15% of all EAC cases are detected by endoscopic surveillance [104]. A ‘liquid biopsy’, such as circulating tumor DNA (ctDNA) and miRNA, has the potential to detect EAC at the early stage without interobserver variability [105]. Sampling biofluid is quicker and less invasive than obtaining cancer tissues, and it is not affected by the anatomical location of the cancer. However, the diagnostic accuracy, cost, and reproducibility must be improved [106,107]. In particular, altered expression of miRNA detected by one method is sometimes not validated by another method. Therefore, careful validation using multiple orthogonal methods is important to check the robustness of findings.
It seems unlikely that a satisfactory detection rate will be achieved by testing for just one miRNA, as various disease conditions can alter extracellular miRNA profiles in the body. However, by combining multiple miRNAs, disease-specific screening may be possible. At present, several companies worldwide are working to develop extracellular miRNA-based diagnostics. Two case-control studies (NCT02464930 and NCT02812680) are registered, both of which are recruiting about 200 participants. However, no population-based study has been performed. We also reported that several cancer types, such as breast and ovarian cancer, can be diagnosed at the early stage by analyzing serum miRNAs [108,109]. However, we also need to identify the underlying biological mechanisms by which these miRNA profiles change under different disease conditions, a process that will take several years. In terms of the cost, it should not be a prohibitive factor for clinical implementation because, if this technology becomes popular, the cost will fall.

5. Conclusions

Many miRNAs have been identified in the context of BE/EAC diagnosis and prognosis. In particular, several studies identified the same serum miRNAs (miR-21-5p, miR-25-3p, and miR93-5p) as being associated with EAC. However, since these studies are small-scale, and the sample types, method of miRNA analysis, pathological definition of disease, study populations, and outcomes vary, it is hard to identify useful miRNAs. To bring miRNA analysis into clinical practice, we must overcome many challenges, including standardization of methodologies, improvement of diagnostic performance, and cost-effectiveness [107]. Further investigations are warranted to firmly establish extracellular miRNAs as useful clinical biomarkers.

Author Contributions

Conceptualization, J.M.; writing—original draft preparation, K.I.; writing—review and editing, J.M. and T.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement.

Not applicable

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xie, S.H.; Lagergren, J. A global assessment of the male predominance in esophageal adenocarcinoma. Oncotarget 2016, 7, 38876–38883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Pohl, H.; Welch, H.G. The role of overdiagnosis and reclassification in the marked increase of esophageal adenocarcinoma incidence. J. Natl. Cancer Inst. 2005, 97, 142–146. [Google Scholar] [CrossRef] [PubMed]
  3. Coleman, H.G.; Xie, S.H.; Lagergren, J. The Epidemiology of Esophageal Adenocarcinoma. Gastroenterology 2018, 154, 390–405. [Google Scholar] [CrossRef] [PubMed]
  4. Wheeler, J.B.; Reed, C.E. Epidemiology of esophageal cancer. Surg. Clin. N. Am. 2012, 92, 1077–1087. [Google Scholar] [CrossRef] [PubMed]
  5. Arnold, M.; Laversanne, M.; Brown, L.M.; Devesa, S.S.; Bray, F. Predicting the Future Burden of Esophageal Cancer by Histological Subtype: International Trends in Incidence up to 2030. Am. J. Gastroenterol. 2017, 112, 1247–1255. [Google Scholar] [CrossRef] [PubMed]
  6. Hanai, M.; Kusano, M.; Kawamura, O.; Shimoyama, Y.; Maeda, M. Epidemiology of Barrett’s esophagus—Comparison of Japan and the West. Nihon Rinsho 2005, 63, 1333–1339. [Google Scholar] [PubMed]
  7. Nishi, T.; Makuuchi, H.; Ozawa, S.; Shimada, H.; Chino, O. The Present Status and Future of Barrett’s Esophageal Adenocarcinoma in Japan. Digestion 2019, 99, 185–190. [Google Scholar] [CrossRef]
  8. Clark, G.W. Effect of Helicobacter pylori infection in Barrett’s esophagus and the genesis of esophageal adenocarcinoma. World J. Surg. 2003, 27, 994–998. [Google Scholar] [CrossRef]
  9. Chow, W.H.; Blaser, M.J.; Blot, W.J.; Gammon, M.D.; Vaughan, T.L.; Risch, H.A.; Perez-Perez, G.I.; Schoenberg, J.B.; Stanford, J.L.; Rotterdam, H.; et al. An inverse relation between cagA+ strains of Helicobacter pylori infection and risk of esophageal and gastric cardia adenocarcinoma. Cancer Res. 1998, 58, 588–590. [Google Scholar]
  10. Gavin, A.T.; Francisci, S.; Foschi, R.; Donnelly, D.W.; Lemmens, V.; Brenner, H.; Anderson, L.A. Oesophageal cancer survival in Europe: A EUROCARE-4 study. Cancer Epidemiol. 2012, 36, 505–512. [Google Scholar] [CrossRef]
  11. Njei, B.; McCarty, T.R.; Birk, J.W. Trends in esophageal cancer survival in United States adults from 1973 to 2009: A SEER database analysis. J. Gastroenterol. Hepatol. 2016, 31, 1141–1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Launoy, G.; Bossard, N.; Castro, C.; Manfredi, S. Trends in net survival from esophageal cancer in six European Latin countries: Results from the SUDCAN population-based study. Eur. J. Cancer Prev. 2017, 26, S24–S31. [Google Scholar] [CrossRef] [PubMed]
  13. Mukaisho, K.I.; Kanai, S.; Kushima, R.; Nakayama, T.; Hattori, T.; Sugihara, H. Barretts’s carcinogenesis. Pathol. Int. 2019, 69, 319–330. [Google Scholar] [CrossRef] [PubMed]
  14. Thomas, T.; Abrams, K.R.; De Caestecker, J.S.; Robinson, R.J. Meta analysis: Cancer risk in Barrett’s oesophagus. Aliment. Pharmacol. Ther. 2007, 26, 1465–1477. [Google Scholar] [CrossRef]
  15. Souza, R.F.; Krishnan, K.; Spechler, S.J. Acid, bile, and CDX: The ABCs of making Barrett’s metaplasia. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G211–G218. [Google Scholar] [CrossRef] [Green Version]
  16. Savarino, E.; De Bortoli, N.; De Cassan, C.; Della Coletta, M.; Bartolo, O.; Furnari, M.; Ottonello, A.; Marabotto, E.; Bodini, G.; Savarino, V. The natural history of gastro-esophageal reflux disease: A comprehensive review. Dis. Esophagus 2017, 30, 1–9. [Google Scholar] [CrossRef]
  17. Arnal, M.J.; Arenas, Á.F.; Arbeloa, Á.L. Esophageal cancer: Risk factors, screening and endoscopic treatment in Western and Eastern countries. World J. Gastroenterol. 2015, 21, 7933–7943. [Google Scholar] [CrossRef]
  18. Bergman, J.; De Groof, A.J.; Pech, O.; Ragunath, K.; Armstrong, D.; Mostafavi, N.; Lundell, L.; Dent, J.; Vieth, M.; Tytgat, G.N.; et al. An Interactive Web-Based Educational Tool Improves Detection and Delineation of Barrett’s Esophagus-Related Neoplasia. Gastroenterology 2019, 156, 1299–1308. [Google Scholar] [CrossRef] [Green Version]
  19. Van der Sommen, F.; Zinger, S.; Curvers, W.L.; Bisschops, R.; Pech, O.; Weusten, B.L.; Bergman, J.J.; De With, P.H.; Schoon, E.J. Computer-aided detection of early neoplastic lesions in Barrett’s esophagus. Endoscopy 2016, 48, 617–624. [Google Scholar] [CrossRef] [Green Version]
  20. Benaglia, T.; Sharples, L.D.; Fitzgerald, R.C.; Lyratzopoulos, G. Health benefits and cost effectiveness of endoscopic and nonendoscopic cytosponge screening for Barrett’s esophagus. Gastroenterology 2013, 144, 62–73. [Google Scholar] [CrossRef]
  21. Kosaka, N.; Iguchi, H.; Ochiya, T. Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 2010, 101, 2087–2092. [Google Scholar] [CrossRef] [PubMed]
  22. Bansal, A.; Hong, X.; Lee, I.H.; House, J.; Mathur, S.C.; Rastogi, A.; Sharma, P.; Christenson, L.K. Serum exosomal microrna expression can be a novel non-invasive strategy for the screening of barrett’s esophagus. Gastroenterology 2013, 144, S-684. [Google Scholar] [CrossRef]
  23. Bus, P.; Kestens, C.; Ten Kate, F.J.W.; Peters, W.; Drenth, J.P.H.; Roodhart, J.M.L.; Siersema, P.D.; Van Baal, J.W.P.M. Profiling of circulating microRNAs in patients with Barrett’s esophagus and esophageal adenocarcinoma. J. Gastroenterol. 2016, 51, 560–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Cabibi, D.; Caruso, S.; Bazan, V.; Castiglia, M.; Bronte, G.; Ingrao, S.; Fanale, D.; Cangemi, A.; Calò, V.; Listì, A.; et al. Analysis of tissue and circulating microRNA expression during metaplastic transformation of the esophagus. Oncotarget 2016, 7, 47821–47830. [Google Scholar] [CrossRef] [Green Version]
  25. Fassan, M.; Saraggi, D.; Balsamo, L.; Realdon, S.; Scarpa, M.; Castoro, C.; Coati, I.; Salmaso, R.; Farinati, F.; Guzzardo, V.; et al. Early miR-223 Upregulation in Gastroesophageal Carcinogenesis. Am. J. Clin. Pathol. 2017, 147, 301–308. [Google Scholar] [CrossRef] [Green Version]
  26. Pavlov, K.; Kluiver, J.; Meijer, C.; Boersma-van Ek, W.; Kruyt, F.A.E.; Karrenbeld, A.; Kleibeuker, J.H.; Peters, F.T.M.; Van den Berg, A. Circulating miRNAs in patients with Barrett’s esophagus, high-grade dysplasia and esophageal adenocarcinoma. J. Gastrointest. Oncol. 2018, 9, 1150–1156. [Google Scholar] [CrossRef] [Green Version]
  27. Wang, L.; Ji, F.; Liu, G.; Wang, W.; Li, Z.; Yue, Y.; Wang, Z. Upregulation of circulating mir130a is correlated with development of barrett’s esophagus and esophageal adenocarcinoma. Onco Targets Ther. 2019, 12, 1–7. [Google Scholar] [CrossRef] [Green Version]
  28. Van Baal, J.W.; Bus, P.; Kestens, C.; Ten Kate, F.T.; Peters, W.H.; Drenth, J.P.; Roodhart, J.; Voest, E.E.; Siersema, P.D. Comprehensive profiling of plasma microRNAs reveals potential biomarkers for barrett’s esophagus and esophageal adenocarcinoma. Gastroenterology 2014, 146, S-97. [Google Scholar] [CrossRef]
  29. Song, J.H.; Cheng, Y.; Yang, J.; Wu, W.; Mori, Y.; Olaru, A.V.; Bootwalla, M.; Sharpf, R.; Meltzer, S.J. Circulating miRNAs as noninvasive biomarkers in esophageal adenocarcinoma patients. Cancer Res. 2011, 71, AM2011–AM4960. [Google Scholar]
  30. Xie, Z.; Chen, G.; Zhang, X.; Li, D.; Huang, J.; Yang, C.; Zhang, P.; Qin, Y.; Duan, Y.; Gong, B.; et al. Salivary microRNAs as promising biomarkers for detection of esophageal cancer. PLoS ONE 2013, 8, e57502. [Google Scholar] [CrossRef]
  31. Chiam, K.; Wang, T.; Watson, D.I.; Mayne, G.C.; Irvine, T.S.; Bright, T.; Smith, L.; White, I.A.; Bowen, J.M.; Keefe, D.; et al. Circulating Serum Exosomal miRNAs As Potential Biomarkers for Esophageal Adenocarcinoma. J. Gastrointest. Surg. 2015, 19, 1208–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Warnecke-Eberz, U.; Chon, S.H.; Hölscher, A.H.; Drebber, U.; Bollschweiler, E. Exosomal onco-miRs from serum of patients with adenocarcinoma of the esophagus: Comparison of miRNA profiles of exosomes and matching tumor. Tumor Biol. 2015, 36, 4643–4653. [Google Scholar] [CrossRef] [PubMed]
  33. Yan, R. Participation of microRNAs in exosome-mediated dysplastic phenotype induction. J. Gastroenterol. Hepatol. 2016, 31, 10. [Google Scholar] [CrossRef]
  34. Zhang, K.; Wu, X.; Wang, J.; Lopez, J.; Zhou, W.; Yang, L.; Wang, S.E.; Raz, D.J.; Kim, J.Y. Circulating miRNA profile in esophageal adenocarcinoma. Am. J. Cancer Res. 2016, 6, 2713–2721. [Google Scholar] [PubMed]
  35. Chen, H. Serum exosomal mirnas expression as novel biomarkers for detection of esophageal adenocarcinoma. United Eur. Gastroenterol. J. 2017, 5, A340. [Google Scholar] [CrossRef]
  36. Matsuzaki, J.; Suzuki, H. Circulating micro RNAs as potential biomarkers to detect transformation of Barrett’s oesophagus to oesophageal Adenocarcinoma. BMJ Open Gastroenterol. 2017, 4. [Google Scholar] [CrossRef] [Green Version]
  37. Miyoshi, J.; Gao, F.; Toden, S.; Shrubsole, M.J.; Scarpa, M.; Whitsett, T.; Inge, L.; Meltzer, S.J.; Teodoro, B.; Coleman, H.G.; et al. A novel non-invasive circulating mirna signature for detection of esophageal adenocarcinoma. Gastroenterology 2018, 154, S-87. [Google Scholar] [CrossRef]
  38. Craig, M.P.; Rajakaruna, S.; Paliy, O.; Sajjad, M.; Madhavan, S.; Reddy, N.; Zhang, J.; Bottomley, M.; Agrawal, S.; Kadakia, M.P. Differential MicroRNA signatures in the pathogenesis of barrett’s esophagus. Clin. Transl. Gastroenterol. 2020, 11, e00125. [Google Scholar] [CrossRef]
  39. Fassan, M.; Realdon, S.; Cascione, L.; Hahne, J.C.; Munari, G.; Guzzardo, V.; Arcidiacono, D.; Lampis, A.; Brignola, S.; Dal Santo, L.; et al. Circulating microRNA expression profiling revealed miR-92a-3p as a novel biomarker of Barrett’s carcinogenesis. Pathol. Res. Pract. 2020, 216, 152907. [Google Scholar] [CrossRef]
  40. Gu, J.; Zhang, J.; Zheng, L.; Ajani, J.A.; Wu, X.; Ye, Y. Serum miR-331-3p predicts tumor recurrence in esophageal adenocarcinoma. Sci. Rep. 2018, 8, 14006. [Google Scholar] [CrossRef]
  41. Odenthal, M.; Hee, J.; Gockel, I.; Sisic, L.; Schmitz, J.; Stoecklein, N.H.; Driemel, C.; Möhlendick, B.; Schmidt, T.; Knoefel, W.T.; et al. Serum microRNA profiles as prognostic/predictive markers in the multimodality therapy of locally advanced adenocarcinomas of the gastroesophageal junction. Int. J. Cancer 2015, 137, 230–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Pu, X.; Gu, J.; Ye, Y.; Ajani, J.; Wu, X. Circulating miRNAs as potential biomarkers for esophageal adenocarcinoma risk and clinical outcomes. Cancer Res. 2014, 74, AM2014–AM2915. [Google Scholar]
  43. Zhai, R.; Wei, Y.; Su, L.; Liu, G.; Kulke, M.H.; Wain, J.C.; Christiani, D.C. Whole-miRNome profiling identifies prognostic serum miRNAs in esophageal adenocarcinoma: The influence of Helicobacter pylori infection status. Carcinogenesis 2015, 36, 87–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Petrick, J.L.; Liao, L.; Wu, X.; Gammon, M.; Vaughan, T.; Cook, M.B. Circulating micrornas in relation to esophageal adenocarcinoma development and survival. Gastroenterology 2018, 154, S-987. [Google Scholar] [CrossRef]
  45. Ergun, S.; Güney, S.; Temiz, E.; Petrovic, N.; Gunes, S. Significance of miR-15a-5p and CNKSR3 as Novel Prognostic Biomarkers in Non-Small Cell Lung Cancer. Anticancer Agents Med. Chem. 2018, 18, 1695–1701. [Google Scholar] [CrossRef] [PubMed]
  46. Kontos, C.K.; Tsiakanikas, P.; Avgeris, M.; Papadopoulos, I.N.; Scorilas, A. miR-15a-5p, A Novel Prognostic Biomarker, Predicting Recurrent Colorectal Adenocarcinoma. Mol. Diagn. Ther. 2017, 21, 453–464. [Google Scholar] [CrossRef]
  47. Wang, Z.M.; Wan, X.H.; Sang, G.Y.; Zhao, J.D.; Zhu, Q.Y.; Wang, D.M. miR-15a-5p suppresses endometrial cancer cell growth via Wnt/β-catenin signaling pathway by inhibiting WNT3A. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4810–4818. [Google Scholar]
  48. Zhao, X.Q.; Tang, H.; Yang, J.; Gu, X.Y.; Wang, S.M.; Ding, Y. MicroRNA-15a-5p down-regulation inhibits cervical cancer by targeting TP53INP1 in vitro. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8219–8229. [Google Scholar] [CrossRef]
  49. Dai, X.; Guo, X.; Liu, J.; Cheng, A.; Peng, X.; Zha, L.; Wang, Z. Circular RNA circGRAMD1B inhibits gastric cancer progression by sponging miR-130a-3p and regulating PTEN and p21 expression. Aging (Albany N. Y.) 2019, 11, 9689–9708. [Google Scholar] [CrossRef]
  50. Kong, X.; Zhang, J.; Li, J.; Shao, J.; Fang, L. MiR-130a-3p inhibits migration and invasion by regulating RAB5B in human breast cancer stem cell-like cells. Biochem. Biophys. Res. Commun. 2018, 501, 486–493. [Google Scholar] [CrossRef]
  51. Shi, H.; Shen, H.; Xu, J.; Zhao, S.; Yao, S.; Jiang, N. MiR-143-3p suppresses the progression of ovarian cancer. Am. J. Transl. Res. 2018, 10, 866–874. [Google Scholar] [PubMed]
  52. Xia, C.; Yang, Y.; Kong, F.; Kong, Q.; Shan, C. MiR-143-3p inhibits the proliferation, cell migration and invasion of human breast cancer cells by modulating the expression of MAPK7. Biochimie 2018, 147, 98–104. [Google Scholar] [CrossRef]
  53. Xie, F.; Li, C.; Zhang, X.; Peng, W.; Wen, T. MiR-143-3p suppresses tumorigenesis in pancreatic ductal adenocarcinoma by targeting KRAS. Biomed. Pharmacother. 2019, 119, 109424. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, K.; Han, B.; Bai, Y.; Ma, X.Y.; Ji, Z.N.; Xiong, Y.; Miao, S.K.; Zhang, Y.Y.; Zhou, L.M. MiR-451a suppressing BAP31 can inhibit proliferation and increase apoptosis through inducing ER stress in colorectal cancer. Cell Death Dis. 2019, 10, 152. [Google Scholar] [CrossRef]
  55. Li, Z.; Ying, X.; Chen, H.; Ye, P.; Shen, Y.; Pan, W.; Zhang, L. MicroRNA-194 inhibits the epithelial-mesenchymal transition in gastric cancer cells by targeting FoxM1. Dig. Dis. Sci. 2014, 59, 2145–2152. [Google Scholar] [CrossRef]
  56. Li, T.T.; Gao, X.; Gao, L.; Gan, B.L.; Xie, Z.C.; Zeng, J.J.; Chen, G. Role of upregulated miR-136-5p in lung adenocarcinoma: A study of 1242 samples utilizing bioinformatics analysis. Pathol. Res. Pract. 2018, 214, 750–766. [Google Scholar] [CrossRef]
  57. Chen, P.; Zhao, L.; Pan, X.; Jin, L.; Lin, C.; Xu, W.; Xu, J.; Guan, X.; Wu, X.; Wang, Y.; et al. Tumor suppressor microRNA-136-5p regulates the cellular function of renal cell carcinoma. Oncol. Lett. 2018, 15, 5995–6002. [Google Scholar] [CrossRef] [Green Version]
  58. Phatak, P.; Burrows, W.M.; Chesnick, I.E.; Tulapurkar, M.E.; Rao, J.N.; Turner, D.J.; Hamburger, A.W.; Wang, J.Y.; Donahue, J.M. MiR-199a-3p decreases esophageal cancer cell proliferation by targeting p21 activated kinase 4. Oncotarget 2018, 9, 28391–28407. [Google Scholar] [CrossRef]
  59. Li, L.; Mou, Y.P.; Wang, Y.Y.; Wang, H.J.; Mou, X.Z. miR-199a-3p targets ETNK1 to promote invasion and migration in gastric cancer cells and is associated with poor prognosis. Pathol. Res. Pract. 2019, 215, 152511. [Google Scholar] [CrossRef]
  60. Fassan, M.; Volinia, S.; Palatini, J.; Pizzi, M.; Fernandez-Cymering, C.; Balistreri, M.; Realdon, S.; Battaglia, G.; Souza, R.; Odze, R.D.; et al. MicroRNA Expression Profiling in the Histological Subtypes of Barrett’s Metaplasia. Clin. Transl. Gastroenterol. 2013, 4, e34. [Google Scholar] [CrossRef]
  61. Vychytilova-Faltejskova, P.; Merhautova, J.; Machackova, T.; Gutierrez-Garcia, I.; Garcia-Solano, J.; Radova, L.; Brchnelova, D.; Slaba, K.; Svoboda, M.; Halamkova, J.; et al. MiR-215-5p is a tumor suppressor in colorectal cancer targeting EGFR ligand epiregulin and its transcriptional inducer HOXB9. Oncogenesis 2017, 6, 399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Gao, J.B.; Zhu, M.N.; Zhu, X.L. miRNA-215-5p suppresses the aggressiveness of breast cancer cells by targeting Sox9. FEBS Open Bio 2019, 9, 1957–1967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Drahos, J.; Schwameis, K.; Orzolek, L.D.; Hao, H.; Birner, P.; Taylor, P.R.; Pfeiffer, R.M.; Schoppmann, S.F.; Cook, M.B. MicroRNA Profiles of Barrett’s Esophagus and Esophageal Adenocarcinoma: Differences in Glandular Non-native Epithelium. Cancer Epidemiol. Biomarkers Prev. 2016, 25, 429–437. [Google Scholar] [CrossRef] [Green Version]
  64. Wei, H.; Cui, R.; Bahr, J.; Zanesi, N.; Luo, Z.; Meng, W.; Liang, G.; Croce, C.M. miR-130a Deregulates PTEN and Stimulates Tumor Growth. Cancer Res. 2017, 77, 6168–6178. [Google Scholar] [CrossRef] [Green Version]
  65. Shen, Y.Y.; Cui, J.Y.; Yuan, J.; Wang, X. MiR-451a suppressed cell migration and invasion in non-small cell lung cancer through targeting ATF2. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 5554–5561. [Google Scholar] [CrossRef]
  66. Liu, J.; Zhu, H.; Yang, X.; Ge, Y.; Zhang, C.; Qin, Q.; Lu, J.; Zhan, L.; Cheng, H.; Sun, X. MicroRNA-21 is a novel promising target in cancer radiation therapy. Tumour Biol. 2014, 35, 3975–3979. [Google Scholar] [CrossRef]
  67. Pfeffer, S.R.; Yang, C.H.; Pfeffer, L.M. The Role of miR-21 in Cancer. Drug Dev. Res. 2015, 76, 270–277. [Google Scholar] [CrossRef]
  68. Jiang, Y.; Chen, X.; Tian, W.; Yin, X.; Wang, J.; Yang, H. The role of TGF-β1-miR-21-ROS pathway in bystander responses induced by irradiated non-small-cell lung cancer cells. Br. J. Cancer 2014, 111, 772–780. [Google Scholar] [CrossRef]
  69. Chen, J.; Gu, Y.; Shen, W. MicroRNA-21 functions as an oncogene and promotes cell proliferation and invasion via TIMP3 in renal cancer. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4566–4576. [Google Scholar]
  70. Masoudi, M.S.; Mehrabian, E.; Mirzaei, H. MiR-21: A key player in glioblastoma pathogenesis. J. Cell. Biochem. 2018, 119, 1285–1290. [Google Scholar] [CrossRef]
  71. Yang, C.H.; Pfeffer, S.R.; Sims, M.; Yue, J.; Wang, Y.; Linga, V.G.; Paulus, E.; Davidoff, A.M.; Pfeffer, L.M. The oncogenic microRNA-21 inhibits the tumor suppressive activity of FBXO11 to promote tumorigenesis. J. Biol. Chem. 2015, 290, 6037–6046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhao, M.Y.; Wang, L.M.; Liu, J.; Huang, X.; Liu, J.; Zhang, Y.F. MiR-21 Suppresses Anoikis through Targeting PDCD4 and PTEN in Human Esophageal Adenocarcinoma. Curr. Med. Sci. 2018, 38, 245–251. [Google Scholar] [CrossRef] [PubMed]
  73. Winther, M.; Alsner, J.; Tramm, T.; Baeksgaard, L.; Holtved, E.; Nordsmark, M. Evaluation of miR-21 and miR-375 as prognostic biomarkers in esophageal cancer. Acta Oncol. 2015, 54, 1582–1591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Xu, X.; Chen, Z.; Zhao, X.; Wang, J.; Ding, D.; Wang, Z.; Tan, F.; Tan, X.; Zhou, F.; Sun, J.; et al. MicroRNA-25 promotes cell migration and invasion in esophageal squamous cell carcinoma. Biochem. Biophys. Res. Commun. 2012, 421, 640–645. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, M.Z.; Cheng, W.C.; Chen, S.F.; Nieh, S.; O’Connor, C.; Liu, C.L.; Tsai, W.W.; Wu, C.J.; Martin, L.; Lin, Y.S.; et al. miR-25/93 mediates hypoxia-induced immunosuppression by repressing cGAS. Nat. Cell Biol. 2017, 19, 1286–1296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Sun, X.Y.; Han, X.M.; Zhao, X.L.; Cheng, X.M.; Zhang, Y. MiR-93-5p promotes cervical cancer progression by targeting THBS2/MMPS signal pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5113–5121. [Google Scholar] [CrossRef]
  77. Matsuzaki, J.; Tsugawa, H.; Masaoka, T.; Mori, H.; Watanabe, M.; Kanai, T.; Suzuki, H. Adiponectin ameliorates carcinogenic pathway depending on miR-221/222 in esophageal adenocarcinoma cells. Gastroenterology 2015, 148, S347. [Google Scholar] [CrossRef]
  78. Chava, S.; Reynolds, C.P.; Pathania, A.S.; Gorantla, S.; Poluektova, L.Y.; Coulter, D.W.; Gupta, S.C.; Pandey, M.K.; Challagundla, K.B. miR-15a-5p, miR-15b-5p, and miR-16-5p inhibit tumor progression by directly targeting MYCN in neuroblastoma. Mol. Oncol. 2020, 14, 180–196. [Google Scholar] [CrossRef] [PubMed]
  79. Sugawara, S.; Yamada, Y.; Arai, T.; Okato, A.; Idichi, T.; Kato, M.; Koshizuka, K.; Ichikawa, T.; Seki, N. Dual strands of the miR-223 duplex (miR-223-5p and miR-223-3p) inhibit cancer cell aggressiveness: Targeted genes are involved in bladder cancer pathogenesis. J. Hum. Genet. 2018, 63, 657–668. [Google Scholar] [CrossRef] [PubMed]
  80. Matsui, D.; Zaidi, A.H.; Martin, S.A.; Omstead, A.N.; Kosovec, J.E.; Huleihel, L.; Saldin, L.T.; DiCarlo, C.; Silverman, J.F.; Hoppo, T.; et al. Primary tumor microRNA signature predicts recurrence and survival in patients with locally advanced esophageal adenocarcinoma. Oncotarget 2016, 7, 81281–81291. [Google Scholar] [CrossRef] [PubMed]
  81. Zhu, X.; Ma, S.P.; Yang, D.; Liu, Y.; Wang, Y.P.; Lin, T.; Li, Y.X.; Yang, S.H.; Zhang, W.C.; Wang, X.L. miR-142-3p Suppresses Cell Growth by Targeting CDK4 in Colorectal Cancer. Cell. Physiol. Biochem. 2018, 51, 1969–1981. [Google Scholar] [CrossRef] [PubMed]
  82. Mansoori, B.; Mohammadi, A.; Ghasabi, M.; Shirjang, S.; Dehghan, R.; Montazeri, V.; Holmskov, U.; Kazemi, T.; Duijf, P.; Gjerstorff, M.; et al. miR-142-3p as tumor suppressor miRNA in the regulation of tumorigenicity, invasion and migration of human breast cancer by targeting Bach-1 expression. J. Cell. Physiol. 2019, 234, 9816–9825. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, X.; Luo, H.; Li, X.; Tian, X.; Peng, B.; Liu, S.; Zhan, T.; Wan, Y.; Chen, W.; Li, Y.; et al. miR-331-3p functions as an oncogene by targeting ST7L in pancreatic cancer. Carcinogenesis 2018, 39, 1006–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Luan, X.; Wang, Y. LncRNA XLOC_006390 facilitates cervical cancer tumorigenesis and metastasis as a ceRNA against miR-331-3p and miR-338-3p. J. Gynecol. Oncol. 2018, 29, e95. [Google Scholar] [CrossRef]
  85. Zhao, D.; Sui, Y.; Zheng, X. MiR-331-3p inhibits proliferation and promotes apoptosis by targeting HER2 through the PI3K/Akt and ERK1/2 pathways in colorectal cancer. Oncol. Rep. 2016, 35, 1075–1082. [Google Scholar] [CrossRef] [Green Version]
  86. Buranjiang, G.; Kuerban, R.; Abuduwanke, A.; Li, X.; Kuerban, G. MicroRNA-331-3p inhibits proliferation and metastasis of ovarian cancer by targeting RCC2. Arch. Med. Sci. 2019, 15, 1520–1529. [Google Scholar] [CrossRef]
  87. Guo, X.; Guo, L.; Ji, J.; Zhang, J.; Zhang, J.; Chen, X.; Cai, Q.; Li, J.; Gu, Q.; Liu, B.; et al. miRNA-331-3p directly targets E2F1 and induces growth arrest in human gastric cancer. Biochem. Biophys. Res. Commun. 2010, 398, 1–6. [Google Scholar] [CrossRef]
  88. Fukumoto, I.; Hanazawa, T.; Kinoshita, T.; Kikkawa, N.; Koshizuka, K.; Goto, Y.; Nishikawa, R.; Chiyomaru, T.; Enokida, H.; Nakagawa, M.; et al. MicroRNA expression signature of oral squamous cell carcinoma: Functional role of microRNA-26a/b in the modulation of novel cancer pathways. Br. J. Cancer 2015, 112, 891–900. [Google Scholar] [CrossRef] [Green Version]
  89. Cao, J.M.; Li, G.Z.; Han, M.; Xu, H.L.; Huang, K.M. MiR-30c-5p suppresses migration, invasion and epithelial to mesenchymal transition of gastric cancer via targeting MTA1. Biomed. Pharmacother. 2017, 93, 554–560. [Google Scholar] [CrossRef]
  90. Song, S.; Long, M.; Yu, G.; Cheng, Y.; Yang, Q.; Liu, J.; Wang, Y.; Sheng, J.; Wang, L.; Wang, Z.; et al. Urinary exosome miR-30c-5p as a biomarker of clear cell renal cell carcinoma that inhibits progression by targeting HSPA5. J. Cell. Mol. Med. 2019, 23, 6755–6765. [Google Scholar] [CrossRef]
  91. Ji, L.; Zhu, Z.N.; He, C.J.; Shen, X. MiR-127-3p targets KIF3B to inhibit the development of oral squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 630–640. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, D.; Tang, L.; Wu, H.; Wang, K.; Gu, D. MiR-127-3p inhibits cell growth and invasiveness by targeting ITGA6 in human osteosarcoma. IUBMB Life 2018, 70, 411–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bi, L.; Yang, Q.; Yuan, J.; Miao, Q.; Duan, L.; Li, F.; Wang, S. MicroRNA-127-3p acts as a tumor suppressor in epithelial ovarian cancer by regulating the BAG5 gene. Oncol. Rep. 2016, 36, 2563–2570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Cao, X.C.; Yu, Y.; Hou, L.K.; Sun, X.H.; Ge, J.; Zhang, B.; Wang, X. miR-142-3p inhibits cancer cell proliferation by targeting CDC25C. Cell Prolif. 2016, 49, 58–68. [Google Scholar] [CrossRef] [Green Version]
  95. Ménard, S.; Pupa, S.M.; Campiglio, M.; Tagliabue, E. Biologic and therapeutic role of HER2 in cancer. Oncogene 2003, 22, 6570–6578. [Google Scholar] [CrossRef] [Green Version]
  96. Yuan, R.; Zhang, X.; Fang, Y.; Nie, Y.; Cai, S.; Chen, Y.; Mo, D. mir-127-3p inhibits the proliferation of myocytes by targeting KMT5a. Biochem. Biophys. Res. Commun. 2018, 503, 970–976. [Google Scholar] [CrossRef]
  97. Li, C.; Zheng, X.; Li, W.; Bai, F.; Lyu, J.; Meng, Q.H. Serum miR-486-5p as a diagnostic marker in cervical cancer: With investigation of potential mechanisms. BMC Cancer 2018, 18, 61. [Google Scholar] [CrossRef] [Green Version]
  98. Gao, Z.J.; Yuan, W.D.; Yuan, J.Q.; Yuan, K.; Wang, Y. miR-486-5p functions as an oncogene by targeting PTEN in non-small cell lung cancer. Pathol. Res. Pract. 2018, 214, 700–705. [Google Scholar] [CrossRef]
  99. Tian, F.; Wang, J.; Ouyang, T.; Lu, N.; Lu, J.; Shen, Y.; Bai, Y.; Xie, X.; Ge, Q. MiR-486-5p Serves as a Good Biomarker in Nonsmall Cell Lung Cancer and Suppresses Cell Growth With the Involvement of a Target PIK3R1. Front. Genet. 2019, 10, 688. [Google Scholar] [CrossRef] [Green Version]
  100. Noto, J.M.; Peek, R.M. The role of microRNAs in Helicobacter pylori pathogenesis and gastric carcinogenesis. Front. Cell. Infect. Microbiol. 2011, 1, 21. [Google Scholar] [CrossRef] [Green Version]
  101. Matsushima, K.; Isomoto, H.; Inoue, N.; Nakayama, T.; Hayashi, T.; Nakayama, M.; Nakao, K.; Hirayama, T.; Kohno, S. MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int. J. Cancer 2011, 128, 361–370. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, F.; Liu, J.; Zou, Y.; Jiao, Y.; Huang, Y.; Fan, L.; Li, X.; Yu, H.; He, C.; Wei, W.; et al. MicroRNA-143-3p, up-regulated in H. pylori-positive gastric cancer, suppresses tumor growth, migration and invasion by directly targeting AKT2. Oncotarget 2017, 8, 28711–28724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Weston, A.P.; Badr, A.S.; Topalovski, M.; Cherian, R.; Dixon, A.; Hassanein, R.S. Prospective evaluation of the prevalence of gastric Helicobacter pylori infection in patients with GERD, Barrett’s esophagus, Barrett’s dysplasia, and Barrett’s adenocarcinoma. Am. J. Gastroenterol. 2000, 95, 387–394. [Google Scholar] [CrossRef] [PubMed]
  104. Rubenstein, J.H.; Shaheen, N.J. Epidemiology, Diagnosis, and Management of Esophageal Adenocarcinoma. Gastroenterology 2015, 149, 302–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Egyud, M.; Tejani, M.; Pennathur, A.; Luketich, J.; Sridhar, P.; Yamada, E.; Ståhlberg, A.; Filges, S.; Krzyzanowski, P.; Jackson, J.; et al. Detection of Circulating Tumor DNA in Plasma: A Potential Biomarker for Esophageal Adenocarcinoma. Ann. Thorac. Surg. 2019, 108, 343–349. [Google Scholar] [CrossRef]
  106. Valihrach, L.; Androvic, P.; Kubista, M. Circulating miRNA analysis for cancer diagnostics and therapy. Mol. Aspects Med. 2020, 72, 100825. [Google Scholar] [CrossRef]
  107. Buschmann, D.; Haberberger, A.; Kirchner, B.; Spornraft, M.; Riedmaier, I.; Schelling, G.; Pfaffl, M.W. Toward reliable biomarker signatures in the age of liquid biopsies—How to standardize the small RNA-Seq workflow. Nucleic Acids Res. 2016, 44, 5995–6018. [Google Scholar] [CrossRef]
  108. Shimomura, A.; Shiino, S.; Kawauchi, J.; Takizawa, S.; Sakamoto, H.; Matsuzaki, J.; Ono, M.; Takeshita, F.; Niida, S.; Shimizu, C.; et al. Novel combination of serum microRNA for detecting breast cancer in the early stage. Cancer Sci. 2016, 107, 326–334. [Google Scholar] [CrossRef]
  109. Yokoi, A.; Matsuzaki, J.; Yamamoto, Y.; Yoneoka, Y.; Takahashi, K.; Shimizu, H.; Uehara, T.; Ishikawa, M.; Ikeda, S.I.; Sonoda, T.; et al. Integrated extracellular microRNA profiling for ovarian cancer screening. Nat. Commun. 2018, 9, 4319. [Google Scholar] [CrossRef]
Figure 1. Flow chart showing the selection process.
Figure 1. Flow chart showing the selection process.
Jcm 10 00117 g001
Figure 2. List of dysregulated miRNAs (identified in serum or saliva) that are specific for BE or EAC. Superscript numbers indicate references. Underlined miRNAs were reported in multiple reports. MiRNAs associated with both OS/prognosis and recurrence are shown as a Venn diagram.
Figure 2. List of dysregulated miRNAs (identified in serum or saliva) that are specific for BE or EAC. Superscript numbers indicate references. Underlined miRNAs were reported in multiple reports. MiRNAs associated with both OS/prognosis and recurrence are shown as a Venn diagram.
Jcm 10 00117 g002
Table 3. Functional roles and validated targets of miRNAs associated with BE diagnosis.
Table 3. Functional roles and validated targets of miRNAs associated with BE diagnosis.
miRNAFunctional RoleTargetCancer TypeSample
miR-15-a-5pTumor promoter
Tumor suppressor
TP53INP1 [48]
WNT3A [47]
Cervical cancer
Endometrial Cancer
30 tissue
8 tissue
miR-130a-3pInhibit cell proliferation, migration, and invasionRAB5B [50]
PTEN, p21 [64]
Breast cancer
Gastric cancer
40 tissue
30 tissue
miR-143-3pTumor suppressor TAK1 [51]
MAPK7 [52]
KRAS [53]
Ovarian cancer
Breast cancer
PDAC
4 cell lines
37 tissue
miR-215-5pTumor suppressorEGFR [61]
Sox9 [62]
Colorectal cancer
Breast cancer
252 tissue
39 tissue
miR-194-5pInhibiting cell migration and invasion during cancer progressionFoxM1 [55]
Gastric cancer50 tissue
miR-451aInhibits the migratory and invasive abilities ATF2 [65]NSCLC55 tissue
miR-199a-3pPrognostic indicator
Tumor suppressor
ETNK1 [59]
PAK4 [58]
Gastric cancer
Esophageal cancer
39 tissue
BE, Barrett’s esophagus; PDAC, pancreatic ductal adenocarcinoma; NSCLC, non-small cell lung cancer.
Table 4. Functional roles and validated targets of miRNAs associated with EAC diagnosis.
Table 4. Functional roles and validated targets of miRNAs associated with EAC diagnosis.
miRNAFunctional RoleTargetCancer TypeSample
miR-21-5pPromotes anoikis resistance and metastasisTGFβ1 [68]
TIMP3 [69]
IGFBP3 [67]
FBXO11 [71]
NSCLC
RCC
GBM
104 tissue
miR-25-3pPromote migration invasionCDH1 [74]SCC
miR-93-5pPromote progressionPTEN [75]
THBS2/MMPS [76]
Breast cancer
Cervical cancer
28 tissue
miR-15a-5pTumor suppressor
Promotor
E2F1, MYCN [78]
TP53INP1 [48]
Neuroblastoma
Cervical cancer
30 tissue
NSCLC: non-small-cell lung carcinoma, RCC: renal cell carcinoma, GBM: glioblastoma multiform, SCC: squamous cell carcinoma.
Table 5. Functional roles and validated targets of miRNAs associated with overall survival, prognosis, and recurrence.
Table 5. Functional roles and validated targets of miRNAs associated with overall survival, prognosis, and recurrence.
miRNAFunctional RoleTargetCancer TypeSample
miR-331-3pTumor suppressorHER2 [84,85]
RCC2 [86]
E2F1 [87]
Pancreatic cancer
Colorectal cancer
Cervical cancer
Ovarian cancer
Gastric cancer
44 plasma
29 tissue
20 tissue
miR-26a-5pTumor suppressorTMEM184B [88]OSCC36 tissue
miR-30c-5pTumor suppressorMTA1 [89]
HSPA5 [90]
Gastric cancer
RCC, Bladder cancer,
Prostate cancer
40 tissue
130 urine
miR-127-3pTumor suppressorKIF3B [91]
ITGA6 [92]
BAG5 [93]
OSCC
Osteosarcoma
Ovarian cancer
45 tissue
20 tissue
miR-142-3pTumor suppressorCDK4 [81]
CDC25C [94]
Beach-1 [82]
Colorectal Cancer

Breast Cancer
116 tissue

42 tissue
OSCC: oral squamous cell carcinoma, RCC: renal cell carcinoma.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Inokuchi, K.; Ochiya, T.; Matsuzaki, J. Extracellular miRNAs for the Management of Barrett’s Esophagus and Esophageal Adenocarcinoma: A Systematic Review. J. Clin. Med. 2021, 10, 117. https://doi.org/10.3390/jcm10010117

AMA Style

Inokuchi K, Ochiya T, Matsuzaki J. Extracellular miRNAs for the Management of Barrett’s Esophagus and Esophageal Adenocarcinoma: A Systematic Review. Journal of Clinical Medicine. 2021; 10(1):117. https://doi.org/10.3390/jcm10010117

Chicago/Turabian Style

Inokuchi, Kazumi, Takahiro Ochiya, and Juntaro Matsuzaki. 2021. "Extracellular miRNAs for the Management of Barrett’s Esophagus and Esophageal Adenocarcinoma: A Systematic Review" Journal of Clinical Medicine 10, no. 1: 117. https://doi.org/10.3390/jcm10010117

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

Article Metrics

Back to TopTop