Review Open Access
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Apr 21, 2020; 26(15): 1708-1725
Published online Apr 21, 2020. doi: 10.3748/wjg.v26.i15.1708
Blood-based biomarkers for early detection of esophageal squamous cell carcinoma
Ling-Yu Chu, Jian-Jun Xie, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
Yu-Hui Peng, Xue-Fen Weng, Yi-Wei Xu, Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
Yu-Hui Peng, Xue-Fen Weng, Jian-Jun Xie, Yi-Wei Xu, Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
ORCID number: Ling-Yu Chu (0000-0002-4682-0931); Yu-Hui Peng (0000-0002-1866-4679); Xue-Fen Weng (0000-0002-2253-5968); Jian-Jun Xie (0000-0002-5141-5076); Yi-Wei Xu (0000-0002-8670-592X).
Author contributions: Chu LY collected the data and wrote the manuscript; Peng YH and Weng XF collected the data; Xie JJ and Xu YW supervised the work and revised the manuscript.
Supported by National Natural Science Foundation of China, No. 31600632 and No. 81972801; Natural Science Foundation of Guangdong Province, No. 2018A030307079 and No. 2019A1515011873; Innovative and Strong School Project of Guangdong, No. 2018KTSCX068; Key Disciplinary Project of Clinical Medicine under the Guangdong High-level University Development Program.
Conflict-of-interest statement: The authors have no conflicts of interest to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Yi-Wei Xu, PhD, Associate Senior Technician, Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515041, Guangdong Province, China. yiwei512@126.com
Received: December 25, 2019
Peer-review started: December 25, 2019
First decision: January 16, 2020
Revised: March 13, 2020
Accepted: March 19, 2020
Article in press: March 19, 2020
Published online: April 21, 2020

Abstract

Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor of the digestive system worldwide, especially in China. Due to the lack of effective early detection methods, ESCC patients often present at an advanced stage at the time of diagnosis, which seriously affects the prognosis of patients. At present, early detection of ESCC mainly depends on invasive and expensive endoscopy and histopathological biopsy. Therefore, there is an unmet need for a non-invasive method to detect ESCC in the early stages. With the emergence of a large class of non-invasive diagnostic tools, serum tumor markers have attracted much attention because of their potential for detection of early tumors. Therefore, the identification of serum tumor markers for early detection of ESCC is undoubtedly one of the most effective ways to achieve early diagnosis and treatment of ESCC. This article reviews the recent advances in the discovery of blood-based ESCC biomarkers, and discusses the origins, clinical applications, and technical challenges of clinical validation of various types of biomarkers.

Key Words: Esophageal squamous cell carcinoma, Biomarker, Diagnosis, Blood-based, Autoantibodies, MicroRNA

Core tip: At present, the early detection of esophageal squamous cell carcinoma (ESCC) mainly depends on endoscopy and histopathological biopsy. However, the high cost and invasiveness of endoscopy have limited its use as a tool for screening the general population. Blood tests provide a non-invasive method for early detection of ESCC. Therefore, this article reviews the recent advances in the discovery of blood-based biomarkers in the early detection of ESCC.



INTRODUCTION

Esophageal cancer (EC) is one of the most aggressive carcinomas of the digestive tract. The incidence of EC ranks seventh among all malignant tumors, and the mortality rate ranks sixth among cancer-related deaths. Globally, there were an estimated 572034 new cases of and 508585 deaths due to EC in 2018[1]. There are two main histological forms of EC: Esophageal adenocarcinoma and esophageal squamous cell carcinoma (ESCC)[2,3]. These two major histological subtypes differ in etiology and geographic incidence, with esophageal adenocarcinoma being more common in Western populations[3-5] and ESCC being more common in the Eastern populations[3,6,7]. China has one of the highest incidences of ESCC, with more than 90% of EC patients in China suffering from ESCC[1,6,7]. In recent years, although the incidence of EC in China has declined, the absolute incidence of EC remains high because of the large population[8].

At present, the five-year survival of EC is 15%-25%[9,10], but survival can be as high as 80% if EC is caught in the early stages[11-13]. The reality is that most EC patients are diagnosed and treated in the advanced stages, which is the primary cause of the poor prognosis of EC. Although endoscopy has been proven to be effective in detecting early-stage EC and can reduce the mortality[14], the high cost and invasiveness limit its use as a wide-ranging screening tool for early-stage EC[15,16] (Table 1). Finding a non-invasive method for early detection of EC will undoubtedly be an effective way to improve the early diagnosis rate and prognosis of EC[16-18].

Table 1 Technologies for detection of esophageal squamous cell carcinoma.
TechnologyQuantitativeQualitativeAdvantagesDisadvantageRef.
Endoscopy-+(1) Obvious observation of esophageal mucosal changes, lesion changes, and lesion size and morphology; and (2) Low false negative rate and false positive rate(1) Invasiveness; (2) Easy to cause complications such as sore throat after examination; and (3) High cost[15,16]
Blood-based biomarker++(1) Non-invasive; (2) Easy to operate; (3) Low cost; (4) Suitable as a screening tool; and (5) Identification of asymptomatic patients at riskHigh false Negative rate and false positive rate[16-18]

Blood-based biomarker tests provide simpler, less invasive alternatives[17]. Early detection of susceptible populations by detecting nucleic acid or protein molecular markers in the blood has become an area of intense investigation in current tumor diagnosis research. In recent years, studies on ESCC serum biomarkers have revealed a variety of cancer-related molecules, including autoantibodies against various tumor-associated antigens (TAAs), microRNAs (miRNAs), various non-coding RNAs, cytokines, proteins, circulating tumor cells (CTC), and circulating tumor DNA (ctDNA). Each type of biomarker provides different information on disease status, with different advantages and disadvantages and different clinical applications. Detection of these biomarkers may provide a new effective means for screening, diagnosis, monitoring, and prognosis of tumors. Here, we provide an overview of the most promising blood-based biomarkers for future screening of ESCC, and extract basic performance characteristics [e.g., sensitivity, specificity, and area under the receiver operating characteristic curve (AUC)] for each study of serum tumor biomarkers.

ORIGIN OF AND SCREENING CRITERIA FOR TUMOR MARKERS

A tumor marker (TM) is defined here as a substance secreted by or released from tumor cells, or a host reaction to the tumor tissue, and present within body fluid and tissue. TMs can reflect the occurrence, development, and detection of tumor response to treatment, and include a wide range of molecules, such as protein, miRNA, RNA, DNA, methylated DNA, metabolites, carbohydrates, autoantibody, lipids, and circulating tumor cells themselves[19]. Since Henry Bence Jones discovered Bence Jones protein in 1846, providing the first TM for clinical diagnosis, in this case for multiple myeloma[20], TMs have been studied for more than 100 years. However, it was not until Abelev et al[21] discovered alpha-fetoprotein in 1963, and Gold and Freeman[22] discovered carcinoembryonic antigen in 1965 that TM assays became widely used clinically.

The advantage of TM detection is that it might be able to demonstrate the existence of a malignant tumor before observable imaging changes, and assist in diagnosis and analysis of the patient condition, leading to the early diagnosis and treatment of cancer. The ideal TM needs to have the following characteristics: (1) High sensitivity and expression in early-stage tumors[23]; (2) Good specificity, only being positive in tumor patients, for differential diagnosis between benign and malignant tumors[23]; (3) The ability to locate the tumor and possess organ specificity (although so far, no markers with complete organ and tumor specificity have been found. At present, alpha-fetoprotein is the only marker that can be used for early diagnosis and screening of primary liver cancer[24], whereas pathological diagnosis is still the main way to diagnose other malignant tumors); (4) Be related to the severity of the disease, tumor size, or stage; (5) Can predict the prognosis of tumor treatment (such as response to postoperative radiotherapy or chemotherapy), and the corresponding TM can be quickly reduced to normal levels and maintained, indicating a good prognosis; for example, prostate-specific antigen (PSA) is a typical biomarker for recurrence after non-surgical treatment of prostate cancer[25]); and (6) Appears in body fluids, especially blood, and is easy to detect.

The research and application of TMs have opened up a new field for tumor diagnosis and clinical treatment. When the amount of tumor-related substances in the body fluid of tumor patients changes, it can indicate the existence of certain tumors. Identifying cancer specific molecules that help to distinguish between normal and cancerous conditions may develop more effective ESCC diagnostic tools.

CANCER-ASSOCIATED AUTOANTIBODIES

The theory of cancer autoantibody production is complex and not fully understood[26]. Studies have shown that the human immune system can sense TAAs with abnormal structure, distribution, and function of certain cellular components involved in tumorigenesis[27], and induce an autoantibody response. In addition, circulating anti-TAA antibodies can be detected several years before clinical diagnosis and might serve as new screening markers[28-30]. Research on autoantibodies has been carried out for more than 100 years[31]. At the beginning, researchers only found that autoantibodies were closely related to autoimmune diseases[32]. However, a large body of epidemiological studies in recent years have shown that patients with autoimmune diseases have a significantly increased or decreased risk for certain cancers, suggesting that autoantibodies may promote or inhibit cancer progression[32,33]. Antigen changes in cancer cells, which have been shown to be closely related to tumor proliferation and grade, induce the production of autoantibodies by the immune system[34]. Therefore, autoantibodies targeting TAAs have been extensively studied in different types of cancer as novel tumor biomarkers. Autoantibodies are not only more sensitive and specific than antigens, but they have been present in all tumor types to date[35,36] and can persist in the serum of cancer patients. With improvements in antibody detection technologies and detection limits, there will be growing utility for autoantibodies as diagnostic biomarkers for ESCC.

Increasing evidence shows that a single cancer-associated autoantibody biomarker has limited diagnostic value. The 70 studies summarized by Xu et al[37] reported that the sensitivity of 49 autoantibodies ranged from 3.9% to 93.7%, with a specificity range of 78.7%-100%. Among the most studied individual markers in ESCC, there are autoantibodies against well-known TAAs such as p53, p16, c-Myc, survivin, NY-ESO-1, and Hsp70 (Table 2)[37-61]. The most commonly used antibody detection method is the ELISA. A meta-analysis by Zhang et al[61] summarized the diagnostic value of anti-p53 for EC, and found that the overall sensitivity and specificity of p53 autoantibodies to EC were 29.6% and 97.9%, respectively. Other studies have shown that anti-p16, c-Myc, survivin, and NY-ESO-1 autoantibodies have a high specificity but poor sensitivity (Table 2). Although one study reported that the sensitivity and specificity of Hsp70 autoantibodies could be as high as 93.7% and 100%, respectively, there were large-scale fluctuations between small samples and different studies[60]. Overall, most anti-TAA autoantibody biomarkers are relatively less sensitive but more specific, indicating limited clinical utility for a single autoantibody. In general, shifting the cutoff toward a higher sensitivity leads to a reduced specificity and vice versa[62].

Table 2 Diagnostic performance of tumor-associated autoantibodies in esophageal squamous cell carcinoma.
Target antigen of autoanti-bodyRef.Year of publicationESCC, nControls, nAll stages/early stage
P valueDetection method
Sensitivity (%)Specificity (%)AUC
p53Zhang et al[38]2016324 (Training)324 (Training)55.9/-89.5/-0.784/-< 0.001ELISA
186 (Validation)186 (Validation)< 0.001ELISA
Xu et al[39]2014388 (Test)125 (Test)30.0/-98.0/-< 0.0001ELISA
237 (Validation)134 (Validation)29.0/-97.0/-< 0.0001ELISA
Qin et al[40]201417424221.8/-96.3/-0.6/-< 0.05ELISA
Chai et al[41]20141578522.9/-100/-< 0.01ELISA
Zhou et al[42]20148820022.0/-98.0/-< 0.01ELISA
Cai et al[43]2008463039.1/22.2100/100< 0.001ELISA
Looi et al[44]200671827.0/-98.8/-< 0.05ELISA
Müller et al[45]20065043620.0/-100/-< 0.05Western blot
Megliorino et al[46]2005778214.3/-97.6/-< 0.01ELISA
Shimada et al[47]200330120530.0/-95.5/-< 0.05ELISA
Shimada et al[48]200210515326.7/20.095.5/95.5< 0.001ELISA
Ralhan et al[49]2000605060.0/-92.0/-< 0.05ELISA
Shimada et al[50]2000356940.0/-100/-< 0.001ELISA
Hagiwara et al[51]2000461328.0/28.6100/100< 0.05ELISA
Shimada et al[52]19985720858.0/-99.0/-< 0.05ELISA
Sobti et al[53]1998202030.0/-100/-0.02ELISA
Cawley et al[54]1998231934.8/-94.7/-0.037ELISA
p16Zhang et al[38]2016324 (Training)324 (Training)29.3/-81.8/-0.60/-< 0.001ELISA
186 (Validation)186 (Validation)< 0.01
Jin et al[55]2015882080.05ELISA
Qin et al[40]201417424218.4/-98.8/-0.6/-< 0.05ELISA
Zhou et al[42]20148820011.0/-97.0/-0.004ELISA
Looi et al[44]2006718214.1/-98.8/-< 0.05ELISA
c-MycZhang et al[38]2016324 (Training)324 (Training)49.1/-81.5/-0.699/-< 0.001ELISA
186 (Validation)186 (Validation)< 0.001ELISA
Qin et al[40]201417424215.5/-98.8/-0.6/-< 0.05ELISA
Zhou et al[42]20148820018.0/-96.0/-< 0.001ELISA
Looi et al[44]200671827.0/-100/-< 0.05ELISA
Megliorino et al[46]2005778211.7/-100/-< 0.01ELISA
SurvivinXiu et al[56]201815936214.5/-90.0/-0.327/-0.524ELISA
Qin et al[40]201417424212.1/-99.6/-< 0.05ELISA
Zhou et al[42]2014882009.0/-96.0/-0.06ELISA
Megliorino et al[46]2005778210.4/-97.6/-< 0.05ELISA
NY-ESO-1Oshima et al[57]20161727432.0/16.0100/100< 0.001ELISA
Xu et al[39]2014388 (Test)125 (Test)26.0/-100/-< 0.0001ELISA
237 (Validation)134 (Validation)24.0/-99.0/-< 0.0001ELISA
Fujita et al[58]200451293.9/-100/-0.532ELISA
Hsp 70Xu et al[39]2014388 (Test)125 (Test)11.0/-99.0/-< 0.001ELISA
237 (Validation)134 (Validation)8.0/-99.0/-< 0.01ELISA
Zhang et al[59]2011697639.1/-92.3/-> 0.01ELISA
Fujita et al[60]2008161393.7/-100/-< 0.001ELISA

Advances in technology have moved the field from investigations of individual candidate anti-TAA autoantibodies to high-throughput, larger-scale discovery efforts using methods such as serological proteome analysis[63] and protein microarrays for the identification of novel anti-TAA autoantibodies[64]. The emergence of these proteomics approaches has facilitated identification of promising anti-TAA autoantibodies. To the best of our knowledge, eight studies have been published on the diagnostic value of different ESCC-related autoantibody biomarker com-binations[37]. In these studies, the autoantibody signatures were able to distinguish ESCCs from healthy controls with a relatively high specificity and variable sensitivity. The sensitivity of autoantibody combinations ranged from 26.0% to 75.3%, and the specificity ranged from 81.0% to 98.8 %. Xu et al[39] used two independent cohorts to study the combination of p53, NY-ESO-1, MMP-7, Hsp70, PRDX-6, and Bmi-1 autoantibodies with sensitivities/specificities of 57.0%/95.0% and 51.0%/96.0%, respectively. They also identified a simplified group of autoantibodies consisting of four anti-TAAs with a similar sensitivity and specificity in early stage ESCC. Another study reported an analysis of c-Myc, HCCR, p53, p62, IMP-1, and Koc in combination. The results showed that the sensitivity/specificity of this combination for distinguishing ESCC patients from the normal control group in the test and validation groups was 67.9%/86.7% and 67.7%/85.5%, respectively[38]. Similar to the research strategy of Xu et al[39], Zhang et al[38] also identified a restricted panel of four TAAs that gave a similar sensitivity and specificity in early-stage ESCC. However, other than the above two studies, other literature did not report the diagnostic efficacy of the autoantibody panel for early-stage ESCC.

Although the above-mentioned anti-TAA autoantibody panel studies have shown satisfactory diagnostic value, due to the different research backgrounds, case characteristics (e.g., diagnostic stage, tumor histology), cut-off values, and experimental methods, we observed that there were some differences in the diagnostic performance of these markers. Moreover, the age difference between the case and control groups in these studies was often large. It is known that the humoral immune response to self-antigens changes with aging[65]. Therefore, the age imbalance between the cases and controls increases the selection bias and cannot be applied to the screening population. Moreover, current research on autoantibodies is lacking, and the anti-TAA autoantibodies we have identified so far may represent only a small fraction of the potential anti-TAA autoantibodies for diagnosis of ESCC. In fact, the diagnostic results of these biomarkers need to be validated in a larger multicenter cohort and evaluated in screening trials for high-risk populations. Therefore, standardized cross-validation studies are needed to validate and quantify the diagnostic potential of these markers[66].

MICRORNAS

MiRNAs are highly conserved, non-coding single-stranded small RNA molecules encoded by endogenous genes, approximately 20 to 24 nucleotides in length[67]. They can be involved in the regulation of a variety of biological functions, including cell differentiation, apoptosis, proliferation, and metabolism, by regulating the expression of target genes[68]. In 2002, Calin et al[69] found that miRNAs are down-regulated in chronic B-lymphocytic leukemia, which is the first report of a relationship between miRNAs and tumors. It is currently believed that miRNAs mediate post-transcriptional gene expression regulation primarily by promoting both target mRNA degradation and protein translation inhibition. A growing number of studies have shown that different miRNAs play different roles in promoting cancer or tumor suppression, and these abnormally expressed miRNAs can unbalance the expression of oncogenic or suppressor genes in the body, eventually leading to tumor production[70]. MiRNAs not only have abnormal expression in tumor tissues, but also have specific expression in patient serum. Recent studies have shown that tumor-derived miRNAs are resistant to endogenous ribonuclease activity, so it can exist in human serum in a stable form[71]. In addition, serum miRNA expression levels are reproducible and consistent among individuals[72], making them ideal candidates for diagnostic screening in blood. Since Zhang et al[73] first reported serum miRNA levels in ESCC patients in 2010, several studies have investigated the differential expression of circulating miRNAs and explored their potential applications in ESCC[74]. Therefore, circulating miRNA markers may contribute to the early diagnosis of ESCC.

To date, increasing studies have confirmed that c-miRNA can be used as a novel serum molecular marker to help early diagnosis of ESCC. Most of the research has focused on candidate miRNAs selected from prior ESCC tissue analysis, while other researchers used high-throughput technology to analyze miRNAs in the discovery sample datasets, and then performed qRT-PCR in an independent verification dataset to determine the diagnostic value of candidate miRNAs[73]. A review by Yao et al[75] of 33 manuscripts investigated a total of 43 different types of miRNAs in serum of ESCC patients. In these studies, the sensitivity, specificity, and AUC of miRNAs in the diagnosis of ESCC were 55.3%-96.9%, 47.4%-100% and 0.590-0.951, respectively[75]. Among the most studied individual miRNAs in ESCC, there are well-known miRNAs, such as miR-21, miR-223, miR-375, miR-25, and miR-100 (Table 3)[73-88]. Wang et al[76] analyzed the diagnostic value of miR-21 and found that it has a good sensitivity and specificity for ESCC, being 71.0% and 96.9%, respectively. However, the number of ESCC patients included in the study was small, and the lack of validation studies of miR-21 limits extension to the clinic. In the current study, the article describes the analysis of the test and validation groups of serum miRNAs, which can serve as potential diagnostic biomarkers for ESCC[73,87,89,90]. The combination of test cohort and validation cohort significantly improved the robustness of the diagnostic accuracy compared to many previous studies without a validation cohort. For example, the serum level of miR-1322 produced an area under the receiver operating characteristic (ROC) curve of 0.847 (95%CI: 0.795-0.890), which was used to distinguish between ESCC and healthy controls in the test group, and similar results were obtained in the validation group (area under the ROC curve: 0.845; 95%CI: 0.780–0.897)[89].

Table 3 Diagnostic performance of microRNAs in esophageal squamous cell carcinoma.
MicroRNARef.Year of publicationESCC, nControls, nAll stages/early stage
P valueDetection method
Sensitivity (%)Specificity (%)AUC
miR-21Wang et al[76]2018313271.0/-96.9/-0.88/-< 0.001qRT-PCR
Sharma et al[77]2018242183.3/-57.2/-0.692/-0.027qRT-PCR
Zhang et al[78]201812512574.0/-78.0/-0.80/0.86< 0.001RT-qPCR
Lv et al[79]2016126800.796/ 0.8120.021qRT-PCR
Li et al[80]201538190.690/-0.017qRT-PCR
Ye et al[81]201410050/97.0/56.0/0.837< 0.001qRT-PCR
Kurashige et al[82]20127139< 0.001qRT-PCR
Wang et al[83]20121743971.0/-69.2/-0.740/-< 0.001qRT-PCR
Komatsu et al[84]201150200.618/-0.022qRT-PCR
miR-223Zhang et al[78]20171251250.68/-0.68/-0.73/0.83< 0.001RT-qPCR
Zhou et al[85]20171371550.649/-< 0.001RT-qPCR
Wu et al[86]2014194980.734/-0.001RT-qPCR
Wu et al[87]201420 (Test)20 (Validation)0.90/-< 0.001RT-qPCR
63 (Test)63 (Validation)0.77/-< 0.001RT-qPCR
Zhang et al[73]201014910083.2/-83.0/-0.911/-< 0.05RT-qPCR
miR-375Zhang et al[78]20171251250.78/-0.59/-0.69/0.87< 0.001RT-qPCR
Lv et al[79]2016126800.712/ 0.7390.023qRT-PCR
Li et al[80]201538190.921/-< 0.0001qRT-PCR
Wu et al[86]2014194980.720/-0.007RT-qPCR
Komatsu et al[84]201150200.807/-0.005qRT-PCR
miR-25Wang et al[76]2018313271.0/-68.8/-0.72/-< 0.001qRT-PCR
Zhang et al[78]20171251250.54/-0.57/-0.55/-0.025RT-qPCR
Wu et al[87]201420 (Test)20 (Validation)0.94/-< 0.001RT-qPCR
63 (Test)63 (Validation)0.78/-< 0.001RT-qPCR
Wu et al[86]20141949847.1/-71.6/-0.593/-0.009RT-qPCR
Komatsu et al[88]2014205085.0/-86.0/-0.856/-< 0.0001RT-qPCR
miR-100Zhang et al[78]20171251250.58/-0.58/-0.58/0.790.164RT-qPCR
Wu et al[87]201420 (Test)20 (Validation)0.88/-< 0.001RT-qPCR
63 (Test)63 (Validation)0.75/-< 0.001RT-qPCR
Zhang et al[73]201014910063.8/-81.0/-0.817/-< 0.05RT-qPCR

Zhang et al[73] measured the serum miRNA concentration by RT-qPCR and identified seven serum miRNAs (miR-10a, miR-22, miR-100, miR-148b, miR-223, miR-133a, and miR-127-3p) that were significantly up-regulated in the serum of ESCC patients compared to the control group. They showed that the seven-miRNA profile could be used as a biomarker for ESCC and, importantly, that it has the potential to predict early ESCC. In addition, this study demonstrated that the seven-miRNA panel was a more sensitive ESCC marker than traditional carcinoembryonic antigen biomarker. Sudo et al[90] established a diagnostic model for serum miRNAs in 566 ESCC patients and 4965 control patients, the largest study to date in designing ESCC diagnostic models. This article[90] used two independent cohorts to study the diagnostic model consisting of miR-8073, miR-6820-5p, miR-6794-5p, miR-3196, miR-744-5p, and miR-6799-5p. The sensitivities/specificities were 100%/98.0% and 96.0%/98.0%, respectively, with similar diagnostic value in early ESCC. In addition, Li et al[91] reviewed 18 publications and investigated 39 different types of miRNAs in EC patients. The authors reported a relatively high sensitivity and specificity of combined and single miRNA markers, indicative of some value in diagnostic application[91]. The results indicated that individual miRNAs showed no statistically significantly higher accuracy than multiple miRNA panels, which is contrary to some previous studies[91]. However, since only two studies in this article compared panels of multiple miRNAs, this finding may not be sufficient to support such conclusion.

Numerous studies have shown that serum circulating miRNAs have potential clinical application as early tumor diagnostic markers, but further clinical data and mechanistic studies are needed for confirmation. The current understanding of miRNA can be summarized as follows. First, the transcription of one miRNA may require the regulation of multiple miRNAs at the same time. On the other hand, one miRNA may be involved in the regulation of the expression of multiple mRNAs at the same time[92]. Obviously, this makes pathway studies of miRNAs more complicated. Second, the processing and detection methods of serum circulating miRNA still need to be standardized, and the selection of internal parameters needs further verification and unification. Finally, most studies on serum circulating miRNAs, in the early diagnosis of tumors, involve small sample size, single-center studies, whereas large-sample, multicenter, prospective clinical studies are needed.

LONG NON-CODING RNAS

Long non-coding RNAs (lncRNAs) are non-coding RNAs that are greater than 200 bases in length, lack an open reading frame, and so have no protein coding ability[93]. LncRNAs regulate gene expression at various levels (epigenetic, transcriptional, and post-transcriptional). LncRNAs regulate gene expression and function in a manner different from miRNAs, which not only affect the post-transcriptional regulation of protein translation, but also function through a variety of pathways that affect gene transcriptional activity and protein degradation[94,95]. A large body of evidence indicates that lncRNAs exert their cancer-promoting or anti-cancer effects by affecting the proliferation, invasion, metastasis, differentiation, apoptosis, and genomic stability of tumor cells[96]. HOX-transcribed RNA (HOTAIR) is the first long non-coding RNA found to have trans-regulatory effects in primary and metastatic breast cancer[97]. In addition, some studies have found that HOTAIR is also highly expressed in ESCC tissues, and the expression level is inversely correlated with degree of differentiation and positively correlated with TNM stage[98]. In recent years, with the maturity and application of whole genome sequencing and lncRNA chips, more and more lncRNAs have been found in different types of tumors, and are closely related to the occurrence and development of tumors, suggesting that lncRNAs could be used as tumor biomarkers[99].

Previous studies on lncRNAs initially focused on tumor tissue. In recent years, investigators have also studied the expression levels of lncRNAs in serum or plasma of tumor patients, and many studies have shown that lncRNAs can be present in extracellular fluids, including serum, plasma, and other body fluids, although the exact mechanism is unclear[100]. In addition, studies by Arita et al[101] confirmed that lncRNAs can stably exist in circulating blood under certain conditions. Recently, a number of laboratories have proposed a variety of serum or plasma lncRNAs that may be used for early diagnosis and efficacy monitoring of ESCC (Table 4)[102-105]. Wang et al[104] used qRT-PCR to detect HOTAIR in serum of ESCC patients, and found that the expression of HOTAIR is increased in serum of ESCC patients, with an area under the diagnostic curve of 0.793, sensitivity of 56.0%, and specificity of 90.0%. Moreover, the level of HOTAIR decreased in serum after ESCC surgery. These results suggest that serum lncRNA-HOTAIR may be a potential diagnostic molecular marker in ESCC[104]. Some studies show that lncRNAs tested alone or in combination exhibit the same or even higher diagnostic performance than traditional cancer biomarkers. Tong et al[103] found that the levels of three lncRNAs, POU3F3, HNF1A-AS1, and SPRY4-IT1, in plasma of ESCC patients were significantly higher than those of normal controls, among which plasma POU3F3 showed the best diagnostic efficacy (area under the curve of 0.842, sensitivity 72.8%, and specificity 89.4%). It is noteworthy that in 147 ESCC and 123 healthy controls, combined detection of plasma POU3F3 and squamous cell carcinoma antigen (SCCA) showed better diagnostic performance (area under the curve of 0.926, sensitivity of 85.7%, and specificity of 81.4%) and an effective detection of 80.8% of patients with early ESCC, suggesting that the combination of POU3F3 and SCCA may be useful for screening early ESCC[103].

Table 4 Diagnostic performance of long non-coding RNAs in esophageal squamous cell carcinoma.
LncRNARef.Year of publicationESCC, nControls, nAll stages/early stage
P valueDetection method
Sensitivity (%)Specificity (%)AUC
POU3F3Hu et al[102]20162052100.584/-< 0.01qRT-PCR
Tong et al[103]201514712372.8/69.289.4/-0.842/-< 0.001qPCR
HOTAIRWang et al[104]2017502056.0/-90.0/-0.793/-< 0.01qRT-PCR
HNF1A-AS1Tong et al[103]201514712332.7/-0.781/-< 0.001qPCR
SPRY4-IT1Tong et al[103]201514712348.2/-0.800/-< 0.001qPCR
linc00152Hu et al[102]20162052100.698/-< 0.01qRT-PCR
CFLAR-AS1Hu et al[102]20162052100.651/-< 0.01qRT-PCR
PGM5-AS1Zhihua et al[105]201941260.894/-< 0.001qRT-PCR

Circulating lncRNAs are thought to be stable in blood because of encapsulation in microvesicles or exosomes[99,101]. A better understanding of the transport of intracellular and intercellular lncRNAs and the underlying biology of cell-derived lipid vesicles may help to develop biomarkers for the detection of human diseases based on circulating lncRNAs. In addition, the detection of biomolecular markers in peripheral blood has the advantage of easy operation and is minimally invasive. Therefore, we expect that the search for new lncRNAs as molecular diagnostic markers in circulating blood will be a hot scientific issue in the field of biomarker research. In order to introduce circulating lncRNAs into clinical practice, further research and improvement should be carried out in the standardization of sample preparation protocols, the control of endogenous lncRNAs in body fluids, and the unification of extraction methods. The criteria for assessing the quality of lncRNAs and the reliability of qPCR results need to be more accurate and reliable, minimizing selection bias[106]. Most of the current research is designed with small samples and thus lacks realistic clinical application at this point. Therefore, it is necessary to further expand the sample size and combine multi-center clinical validation studies to develop an lncRNA detection kit for marker detection in blood, thereby improving the early diagnosis and postoperative monitoring efficiency of lncRNAs in tumors.

CIRCULATING TUMOR DNA

In 1948, Mandel and Metais first reported the presence of circulating cell free-DNA (cf-DNA) in human peripheral blood[107]. cf-DNA refers to extracellular DNA found in body fluids such as blood, cerebrospinal fluid, and synovial fluid, and is a degradation product of endogenous DNA in cells. In recent decades, many studies have found that cf-DNA levels are higher in cancer patients, especially in the advanced stages[108,109]. Researchers first detected KRAS oncogenic mutations in the blood cf-DNA of patients with pancreatic cancer in 1994 by using PCR, which was consistent with that detected in tumor tissues. In other words, the small part of cf-DNA carrying tumor-specific mutations is indeed released by tumor cells[110]. Thus, tumor-associated mutations in cf-DNA can serve as tumor-specific markers, and these tumor-derived cf-DNA fragments carrying tumor characteristics are referred to as ctDNA[110,111]. ctDNA is DNA fragments released by apoptotic or necrotic cells into the blood vessels, and is mainly present in extracellular plasma[112]. The concentration of ctDNA in advanced tumors is between 0.1% and 10%, and is positively correlated with tumor stage and tumor volume[113]. Because the content of ctDNA in total plasma DNA is small, the detection and quantification of ctDNA are very challenging. At present, the quantitative technology of ctDNA has developed from quantitative polymerase chain reaction to complex BEAMing and deep next-generation sequencing, thereby improving the sensitivity and specificity of ctDNA detection[114]. With the development of sensitive technologies to detect rare mutations, the use of blood samples can determine tumor heterogeneity.

As a new molecular marker for tumors, ctDNA is being studied more and more extensively in the field of tumors. It shows great potential for clinical application in the early diagnosis of tumors, residual and recurrence monitoring, and prognosis, which has brought subversive changes to traditional tumor diagnosis and treatment. In recent years, ctDNA methylation has become a highly sensitive method for detecting landmark characteristics of tumors. Kawakami et al[115] observed that high-level methylation of APC DNA occurs in 61% of ESCC patients, and its high expression is associated with poor prognosis. Moreover, Hibi et al[116] detected abnormal methylation of the driver P16 gene in 18% of ESCC patients. Liu et al[117] evaluated the methylation status of Wnt antagonist family genes in EC patients by applying methylation-specific PCR to detect hypermethylation of the driving factors SFRP-1/WIF-1, DKK-3, and RUNX-3 genes in plasma. Therefore, measuring abnormally high levels of methylation of drivers of cancer-related genes might be used for diagnosis of ESCC and monitoring recurrence. Increasing studies have confirmed that detection of ctDNA in the blood of tumor patients can also identify all driver gene mutations in the tumor tissue[118,119]. Lebofsky et al[120] performed in-depth sequencing analysis of plasma ctDNA and metastatic tumor tissue from 34 tumor patients (including 18 different tumor types), covering 6800 COSMIC tumor hotspot mutations in 46 genes. The results showed that in 27 patients, 28 (97%) of 29 mutant genes in metastatic tumor tissue were detected in paired plasma ctDNA[120]. These results indicate that plasma ctDNA has the potential to replace tumor metastatic lesion tissue for the detection of mutant genes[120,121]. Plasma ctDNA samples are easy to obtain, with good patient dependence, and the operations can be repeated. It is a feasible tumor molecular marker that might replace tissue biopsy for metastatic tumor gene mutation. Compared with tissue biopsy, ctDNA has the advantages of non-invasive operation and providing more comprehensive tumor genomics information[122,123]. Another major clinical application of ctDNA detection is the dynamic monitoring of tumor burden. At the same time, ctDNA detection could detect tumor progression 5 to 10 mo in advance[124,125], and detect disease progression earlier than traditional detection methods. However, the clinical application of ctDNA testing still has the following difficulties: (1) Detection technology is still immature and there is a lack of standardized ctDNA extraction and detection procedures; (2) Testing costs are expensive; and (3) There is a lack of large sample, prospective clinical studies to evaluate the early diagnostic value in cancers. In the future, with the development of gene sequencing technology and precision medicine, the application of ctDNA technology in clinical practice will be just around the corner.

METABOLITES

Metabolomics is an emerging discipline that studies the composition, content changes, and interrelationships of all small molecule metabolites in biological samples at specific times or in given environments. In 1999, Nicholson et al[126] formally put forward the concept of “metabolomics”, the qualitative and quantitative analysis of dynamic changes of all metabolic components (intermediate products and end products) of a biological system under pathophysiological conditions. It is the continuation and development of genomics, transcriptomics, and proteomics, and is at the end of the regulation of life activities[127]. In recent years, related research on metabolomics in tumors has also been given increasing attention. Pathological changes in tumor development often lead to significant changes in basic metabolism, resulting in changes in the relative level of small molecule metabolites, which ultimately show the difference between the metabolic spectrum of tumor patients and that of healthy controls[128]. Metabolomics uses advanced analytical chemistry techniques to comprehensively measure a large number of small molecule metabolites in cells, tissues, and body fluids[129]. At the same time, combined with bioinformatics and other methods, changes in the body's small molecule metabolites are analyzed during tumor development, and a tumor metabolism map is finally drawn[130]. It is well known that small changes in gene and protein levels often lead to significant changes in metabolite levels, so metabolomics is a highly sensitive and direct method of disease detection. In recent years, with the development of metabolomics technology, the diagnosis and prognosis of tumors based on metabolomics analysis have been greatly improved[128].

The study of metabolomics depends on the development of various related analytical chemistry technologies. At present, the spectroscopic techniques of metabolic analysis have been mainly limited to nuclear magnetic resonance and mass spectrometry (MS), the latter requiring a combination of separation techniques, to enable analysis by gas chromatography-MS (GC-MS) or liquid chromatography-MS[128]. Recently, investigators have applied the latest metabolomics techniques to explore abnormal metabolic changes in tumors and found many metabolites that are abnormally elevated in specific tumors, such as glucose, serine, lactic acid, and polyamines[131]. There have been many clinical advances in ESCC based on metabolomics[132,133], most of which are non-targeted metabolomics studies focusing on the identification of diagnostic biomarkers for ESCC, but not prognosis for ESCC metastasis[132]. Jin et al[132] used gas chromatography-MS to measure serum metabolome molecular marker levels in 60 ESCC patients and 30 normal controls. They developed a prediction model consisting of three metabolic molecules, valine, γ-aminobutyric acid, and pyrrole-2-carboxylic acid, which gave an area under the curve, sensitivity, and specificity of 0.964, 90.0%, and 96.67%, respectively. The diagnostic effectiveness of this predictive model was almost same as the validation set. Liu et al[134] conducted metabolomics analysis on the plasma of 53 ESCC patients and matched 53 normal controls, and found that 25 metabolites were up-regulated and 5 metabolites were down-regulated. Subsequent database verification identified 11 metabolites, of which 6 were the phospholipids phosphatidylserine, phosphatidic acid, lecithin, phosphatidylinositol, phosphatidylethanolamine, and sphinganine 1-phosphate. Ma et al[129] applied high performance liquid chromatography to analyze plasma free amino acids in patients with ESCC, and the results showed that there are many differences in plasma free amino acid metabolism profiles between ESCC patients and healthy controls, including Asp, Ala, Glu, Gly, and Thr, suggesting that plasma free amino acids may help distinguish ESCC from healthy controls.

The above-mentioned studies show that high-throughput detection methods of metabolomics can illustrate the whole picture of small molecule metabolic markers in tumor body, thus providing a new way to find ideal molecular markers for the early diagnosis of tumors. However, metabolomics is an emerging discipline, and its development faces many difficulties. First, it is unclear how many metabolites exist in the human metabolome. Second, the various substances produced by human metabolism are complex and involve different biochemical categories. Currently, no platform can achieve comprehensive identification and simultaneous measurement of all metabolites. Finally, the sources of metabolites in human samples are different, and changes in the same metabolite between different individuals will also be affected by multiple factors, which stands as a barrier for the implementation of metabolomics research. Of course, at present, tumor metabolomics is still in the initial exploration stage. In future work, the clinical application of metabolomics still needs more experiments and clinical research for systematic and comprehensive verification.

CYTOKINES

Cytokines are a class of low-molecular-weight soluble substances with high activity and multifunctionality produced by various cells, such as immune cells activated by immunogens, mitogens, or other stimulants, most of which are peptides or small molecular glycoproteins[135,136]. They play a role in intercellular communication and cell growth, and participate in cell differentiation, migration, and apoptosis[137]. These mediators are involved in signal transduction between cells, and regulate the human immune response, promote hematopoietic and anti-inflammatory effects and anti-viral immunity, participate in tumorigenesis and development, and are involved in various pathophysiological processes[135,136]. According to their structure and function, cytokines can be divided into interleukins (ILs) (such as IL-6), interferons, chemokines (such as IL-8), growth factors [such as vascular endothelial growth factor (VEGF)], colony-stimulating factors, and the tumor necrosis factor superfamily. Some inflammatory cytokines are involved in different molecular mechanisms leading to canceration[138]. It is well known that the process of malignant transformation of tumor cells involves the expression and activity of a variety of cytokines. These cytokines play an important role in tumorigenesis, angiogenesis, and induction of metastasis, and are also potential molecular markers for tumor diagnosis.

There is transient overexpression of cytokines in many disease states. In cancer, the changes in the production of cytokines increase with the progression of the disease, and participate in or even promote the progression of tumors. As a result, different cytokines are deregulated, and their altered local and systemic concentrations can be detected in body fluids as biomarkers of cancer. In recent years, more and more cytokines have been confirmed to be abnormally expressed in the serum of ESCC patients[139-147], and may be used as molecular markers for ESCC diagnosis. The ESCC cytokine network is rich in pro-inflammatory cytokines, growth factors, and chemokines. The main ESCC-related cytokines are VEGF-A, VEGF-C, IL-6, and IL-8 (Table 5)[139-147]. Kozłowski et al[139] performed an analysis of 89 ESCC patients and 30 healthy controls and showed that the diagnostic sensitivity, specificity, and AUC of VEGF-A for ESCC were 83.0%, 70.0%, and 0.865, respectively. Another analysis of ESCC[140] showed that IL-8, VEGF-C, and VEGF-A expression levels were significantly higher in 70 ESCC patients than in 42 normal controls. Combining both IL-8 and VEGF-C, the AUC that distinguishes ESCC from normal controls is better than that of IL-8 or VEGF-C tested alone. These results indicate that IL-8 and VEGF-C can potentially be used as cytokine molecular markers for the detection of ESCC. Those authors further analyzed the correlation between IL-8 and VEGF-C and VEGF-A, and found that IL-8 and VEGF-C are more closely related. Therefore, the authors speculated that IL-8 may work by stimulating the expression and secretion of VEGF-C[140]. Łukaszewicz-Zając et al[147] measured the levels of serum IL-6 in 90 healthy controls and 30 ESCC patients, and found that IL-6 levels in ESCC patients were increased compared to the controls. Further ROC curve analysis results showed that the detection sensitivity of IL-6 was 87%, specificity was 92%, and AUC was 0.924, suggesting that IL-6 may be helpful for the diagnosis of ESCC.

Table 5 Diagnostic performance of cytokines in esophageal squamous cell carcinoma.
CytokineRef.Year of publicationESCC, nControls, nAll stages/early stage
P valueDetection method
Sensitivity (%)Specificity (%)AUC
VEGF-AKozłowski et al[139]2013893083.0/-70.0/-0.865/-< 0.001ELISA
Krzystek-Korpacka et al[140]2008704272.5/-66.0/-0.739/-< 0.001ELISA
Krzystek-Korpacka et al[141]2007704770.0/-81.0/-0.837/-< 0.001ELISA
Ren et al[142]20057215< 0.001ELISA
Shimada et al[143]2001962479.0/-48.0/-0.001ELISA
VEGF-CKozlowski et al[144]20101103060.0/-80.0/-< 0.001ELISA
Krzystek-Korpacka et al[140]2008704278.6/-76.6/-0.841/-< 0.001ELISA
IL-8Tong et al[145]20181010< 0.05ELISA
Krzystek-Korpacka et al[140]2008704277.1/-74.4/-0.782/-< 0.001ELISA
Ren et al[146]200514935< 0.001ELISA
Ren et al[142]20057215< 0.001ELISA
IL-6Tong et al[145]20181010< 0.05ELISA
Łukaszewicz-Zając et al[147]2011309087.0/-92.0/-0.924/-< 0.001ELISA

The future potential of cytokines seems to be primarily related to their prognosis and predictive value[139,146]. Cytokines can also be used as markers for monitoring treatment response and disease recurrence[140-142]. At present, significant progress has also been made in exploring cytokines as molecular markers for the early diagnosis of tumors. However, most studies lack the evaluation of early tumor samples or samples before diagnosis. In the future, whether cytokines can be used clinically for early diagnosis of tumors still needs high-quality large samples and prospective studies for further confirmation.

CONCLUSION

Early diagnosis is one of the most effective ways to improve the survival rate and reduce the mortality of cancer patients. Clinically, endoscopy can detect early ESCC and its precancerous lesions. A recent large-scale prospective study confirmed for the first time that esophageal endoscopy screening and intervention can effectively reduce the incidence and mortality of ESCC[14]. However, endoscopy is an invasive diagnostic and screening method, which limits its widespread use in the screening of asymptomatic people, making the development and validation of non-invasive biomarkers important for the screening of ESCC. Although some new serological markers have been studied, these have not been translated into effective clinical tools.

In the field of biomarker research related to ESCC, although many studies have shown that biomarkers have diagnostic potential for early ESCC, on the whole, research on the diagnostic effects of these biomarkers on ESCC still have many limitations, such as small sample size, research design of a single population, lack of value for early diagnosis, lack of independent verification tests, and lack of pre-clinical data. Meanwhile, we note that some of the studies in this review did not include patients with early ESCC, so in future studies, the early diagnostic value of these markers needs to be further evaluated. Moreover, there is insufficient molecular profiling data on potential circulating biomarkers for ESCC diagnosis and prognosis. Therefore, in order to realize the clinical application of autoantibodies and the early diagnosis of ESCC, it is still necessary to further screen and identify biomarkers with better diagnostic efficiency and optimize the best combination. Moreover, results need to be confirmed for large sample sizes in multi-center prospective studies.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): 0

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Otowa Y S-Editor: Dou Y L-Editor: Wang TQ E-Editor: Ma YJ

References
1.  Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53206]  [Cited by in F6Publishing: 50898]  [Article Influence: 8483.0]  [Reference Citation Analysis (44)]
2.  Bandla S, Pennathur A, Luketich JD, Beer DG, Lin L, Bass AJ, Godfrey TE, Litle VR. Comparative genomics of esophageal adenocarcinoma and squamous cell carcinoma. Ann Thorac Surg. 2012;93:1101-1106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
3.  Gupta B, Kumar N. Worldwide incidence, mortality and time trends for cancer of the oesophagus. Eur J Cancer Prev. 2017;26:107-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 136]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
4.  Devesa SS, Blot WJ, Fraumeni JF. Changing patterns in the incidence of esophageal and gastric carcinoma in the United States. Cancer. 1998;83:2049-2053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
5.  Arnold M, Soerjomataram I, Ferlay J, Forman D. Global incidence of oesophageal cancer by histological subtype in 2012. Gut. 2015;64:381-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 944]  [Cited by in F6Publishing: 941]  [Article Influence: 104.6]  [Reference Citation Analysis (0)]
6.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23762]  [Cited by in F6Publishing: 25182]  [Article Influence: 1937.1]  [Reference Citation Analysis (3)]
7.  Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18694]  [Cited by in F6Publishing: 20838]  [Article Influence: 2315.3]  [Reference Citation Analysis (2)]
8.  Li J, Qi Z, Hu YP, Wang YX. Possible biomarkers for predicting lymph node metastasis of esophageal squamous cell carcinoma: a review. J Int Med Res. 2019;47:544-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Pennathur A, Gibson MK, Jobe BA, Luketich JD. Oesophageal carcinoma. Lancet. 2013;381:400-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1629]  [Cited by in F6Publishing: 1800]  [Article Influence: 163.6]  [Reference Citation Analysis (4)]
10.  Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med. 2003;349:2241-2252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2115]  [Cited by in F6Publishing: 2142]  [Article Influence: 102.0]  [Reference Citation Analysis (0)]
11.  Wang J, Wu N, Zheng QF, Yan S, Lv C, Li SL, Yang Y. Evaluation of the 7th edition of the TNM classification in patients with resected esophageal squamous cell carcinoma. World J Gastroenterol. 2014;20:18397-18403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 32]  [Cited by in F6Publishing: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
12.  Bird-Lieberman EL, Fitzgerald RC. Early diagnosis of oesophageal cancer. Br J Cancer. 2009;101:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 88]  [Article Influence: 5.9]  [Reference Citation Analysis (1)]
13.  Jankowski J, Barr H, Wang K, Delaney B. Diagnosis and management of Barrett's oesophagus. BMJ. 2010;341:c4551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
14.  Wei WQ, Chen ZF, He YT, Feng H, Hou J, Lin DM, Li XQ, Guo CL, Li SS, Wang GQ, Dong ZW, Abnet CC, Qiao YL. Long-Term Follow-Up of a Community Assignment, One-Time Endoscopic Screening Study of Esophageal Cancer in China. J Clin Oncol. 2015;33:1951-1957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 214]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
15.  Lao-Sirieix P, Fitzgerald RC. Screening for oesophageal cancer. Nat Rev Clin Oncol. 2012;9:278-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 105]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
16.  Gerson LB, Groeneveld PW, Triadafilopoulos G. Cost-effectiveness model of endoscopic screening and surveillance in patients with gastroesophageal reflux disease. Clin Gastroenterol Hepatol. 2004;2:868-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 90]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
17.  Hinz R, Schwarz NG, Hahn A, Frickmann H. Serological approaches for the diagnosis of schistosomiasis - A review. Mol Cell Probes. 2017;31:2-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 77]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
18.  Wang JY, Hsieh JS, Chang MY, Huang TJ, Chen FM, Cheng TL, Alexandersen K, Huang YS, Tzou WS, Lin SR. Molecular detection of APC, K- ras, and p53 mutations in the serum of colorectal cancer patients as circulating biomarkers. World J Surg. 2004;28:721-726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 116]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
19.  Henry NL, Hayes DF. Cancer biomarkers. Mol Oncol. 2012;6:140-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 425]  [Cited by in F6Publishing: 442]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
20.  Clamp JR. Some aspects of the first recorded case of multiple myeloma. Lancet. 1967;2:1354-1356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 33]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
21.  Abelev GI, Perova SD, Khramkova NI, Postnikova ZA, Irlin IS. Production of embryonal alpha-globulin by transplantable mouse hepatomas. Transplantation. 1963;1:174-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 444]  [Cited by in F6Publishing: 474]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
22.  Gold P, Freedman SO. Demonstration of tumor-specific antigens in human colonic carcinomata by immunological tolerance and absorption techniques. J Exp Med. 1965;121:439-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1572]  [Cited by in F6Publishing: 1505]  [Article Influence: 53.8]  [Reference Citation Analysis (0)]
23.  Holdhoff M, Yovino SG, Boadu O, Grossman SA. Blood-based biomarkers for malignant gliomas. J Neurooncol. 2013;113:345-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
24.  Bruix J, Sherman M, Llovet JM, Beaugrand M, Lencioni R, Burroughs AK, Christensen E, Pagliaro L, Colombo M, Rodé, s J, EASL Panel of Experts on HCC. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL conference. European Association for the Study of the Liver. J Hepatol. 2001;35:421-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3252]  [Cited by in F6Publishing: 3170]  [Article Influence: 137.8]  [Reference Citation Analysis (0)]
25.  Patel DA, Presti JC, McNeal JE, Gill H, Brooks JD, King CR. Preoperative PSA velocity is an independent prognostic factor for relapse after radical prostatectomy. J Clin Oncol. 2005;23:6157-6162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 80]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
26.  Zaenker P, Ziman MR. Serologic autoantibodies as diagnostic cancer biomarkers--a review. Cancer Epidemiol Biomarkers Prev. 2013;22:2161-2181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 109]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
27.  Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science. 2011;331:1565-1570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3828]  [Cited by in F6Publishing: 4144]  [Article Influence: 318.8]  [Reference Citation Analysis (0)]
28.  Chapman CJ, Thorpe AJ, Murray A, Parsy-Kowalska CB, Allen J, Stafford KM, Chauhan AS, Kite TA, Maddison P, Robertson JF. Immunobiomarkers in small cell lung cancer: potential early cancer signals. Clin Cancer Res. 2011;17:1474-1480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 74]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
29.  Zhong L, Coe SP, Stromberg AJ, Khattar NH, Jett JR, Hirschowitz EA. Profiling tumor-associated antibodies for early detection of non-small cell lung cancer. J Thorac Oncol. 2006;1:513-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 48]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
30.  Li Y, Karjalainen A, Koskinen H, Hemminki K, Vainio H, Shnaidman M, Ying Z, Pukkala E, Brandt-Rauf PW. p53 autoantibodies predict subsequent development of cancer. Int J Cancer. 2005;114:157-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
31.  Graham JB, Graham RM. Antibodies elicited by cancer in patients. Cancer. 1955;8:409-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
32.  Wu J, Li X, Song W, Fang Y, Yu L, Liu S, Churilov LP, Zhang F. The roles and applications of autoantibodies in progression, diagnosis, treatment and prognosis of human malignant tumours. Autoimmun Rev. 2017;16:1270-1281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
33.  Bernatsky S, Ramsey-Goldman R, Labrecque J, Joseph L, Boivin JF, Petri M, Zoma A, Manzi S, Urowitz MB, Gladman D, Fortin PR, Ginzler E, Yelin E, Bae SC, Wallace DJ, Edworthy S, Jacobsen S, Gordon C, Dooley MA, Peschken CA, Hanly JG, Alarcón GS, Nived O, Ruiz-Irastorza G, Isenberg D, Rahman A, Witte T, Aranow C, Kamen DL, Steinsson K, Askanase A, Barr S, Criswell LA, Sturfelt G, Patel NM, Senécal JL, Zummer M, Pope JE, Ensworth S, El-Gabalawy H, McCarthy T, Dreyer L, Sibley J, St Pierre Y, Clarke AE. Cancer risk in systemic lupus: an updated international multi-centre cohort study. J Autoimmun. 2013;42:130-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 197]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
34.  Chen Z, Huang X, Ye J, Pan P, Cao Q, Yang B, Li Z, Su M, Huang C, Gu J. Immunoglobulin G is present in a wide variety of soft tissue tumors and correlates well with proliferation markers and tumor grades. Cancer. 2010;116:1953-1963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 46]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
35.  Preuss KD, Zwick C, Bormann C, Neumann F, Pfreundschuh M. Analysis of the B-cell repertoire against antigens expressed by human neoplasms. Immunol Rev. 2002;188:43-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 84]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
36.  Li G, Miles A, Line A, Rees RC. Identification of tumour antigens by serological analysis of cDNA expression cloning. Cancer Immunol Immunother. 2004;53:139-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
37.  Xu YW, Peng YH, Xu LY, Xie JJ, Li EM. Autoantibodies: Potential clinical applications in early detection of esophageal squamous cell carcinoma and esophagogastric junction adenocarcinoma. World J Gastroenterol. 2019;25:5049-5068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
38.  Zhang HF, Qin JJ, Ren PF, Shi JX, Xia JF, Ye H, Wang P, Song CH, Wang KJ, Zhang JY. A panel of autoantibodies against multiple tumor-associated antigens in the immunodiagnosis of esophageal squamous cell cancer. Cancer Immunol Immunother. 2016;65:1233-1242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
39.  Xu YW, Peng YH, Chen B, Wu ZY, Wu JY, Shen JH, Zheng CP, Wang SH, Guo HP, Li EM, Xu LY. Autoantibodies as potential biomarkers for the early detection of esophageal squamous cell carcinoma. Am J Gastroenterol. 2014;109:36-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 80]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
40.  Qin JJ, Wang XR, Wang P, Ren PF, Shi JX, Zhang HF, Xia JF, Wang KJ, Song CH, Dai LP, Zhang JY. Mini-array of multiple tumor-associated antigens (TAAs) in the immunodiagnosis of esophageal cancer. Asian Pac J Cancer Prev. 2014;15:2635-2640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
41.  Chai Y, Peng B, Dai L, Qian W, Zhang Y, Zhang JY. Autoantibodies response to MDM2 and p53 in the immunodiagnosis of esophageal squamous cell carcinoma. Scand J Immunol. 2014;80:362-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
42.  Zhou SL, Yue WB, Fan ZM, Du F, Liu BC, Li B, Han XN, Ku JW, Zhao XK, Zhang P, Cui J, Zhou FY, Zhang LQ, Fan XP, Zhou YF, Zhu LL, Liu HY, Wang LD. Autoantibody detection to tumor-associated antigens of P53, IMP1, P16, cyclin B1, P62, C-myc, Survivn, and Koc for the screening of high-risk subjects and early detection of esophageal squamous cell carcinoma. Dis Esophagus. 2014;27:790-797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
43.  Cai HY, Wang XH, Tian Y, Gao LY, Zhang LJ, Zhang ZY. Changes of serum p53 antibodies and clinical significance of radiotherapy for esophageal squamous cell carcinoma. World J Gastroenterol. 2008;14:4082-4086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
44.  Looi K, Megliorino R, Shi FD, Peng XX, Chen Y, Zhang JY. Humoral immune response to p16, a cyclin-dependent kinase inhibitor in human malignancies. Oncol Rep. 2006;16:1105-1110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
45.  Müller M, Meyer M, Schilling T, Ulsperger E, Lehnert T, Zentgraf H, Stremmel W, Volkmann M, Galle PR. Testing for anti-p53 antibodies increases the diagnostic sensitivity of conventional tumor markers. Int J Oncol. 2006;29:973-980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
46.  Megliorino R, Shi FD, Peng XX, Wang X, Chan EK, Tan EM, Zhang JY. Autoimmune response to anti-apoptotic protein survivin and its association with antibodies to p53 and c-myc in cancer detection. Cancer Detect Prev. 2005;29:241-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
47.  Shimada H, Ochiai T, Nomura F; Japan p53 Antibody Research Group. Titration of serum p53 antibodies in 1,085 patients with various types of malignant tumors: a multiinstitutional analysis by the Japan p53 Antibody Research Group. Cancer. 2003;97:682-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 103]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
48.  Shimada H, Nabeya Y, Okazumi S, Matsubara H, Funami Y, Shiratori T, Hayashi H, Takeda A, Ochiai T. Prognostic significance of serum p53 antibody in patients with esophageal squamous cell carcinoma. Surgery. 2002;132:41-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 42]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
49.  Ralhan R, Arora S, Chattopadhyay TK, Shukla NK, Mathur M. Circulating p53 antibodies, p53 gene mutational profile and product accumulation in esophageal squamous-cell carcinoma in India. Int J Cancer. 2000;85:791-795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
50.  Shimada H, Takeda A, Arima M, Okazumi S, Matsubara H, Nabeya Y, Funami Y, Hayashi H, Gunji Y, Suzuki T, Kobayashi S, Ochiai T. Serum p53 antibody is a useful tumor marker in superficial esophageal squamous cell carcinoma. Cancer. 2000;89:1677-1683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
51.  Hagiwara N, Onda M, Miyashita M, Sasajima K. Detection of circulating anti-p53 antibodies in esophageal cancer patients. J Nippon Med Sch. 2000;67:110-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
52.  Shimada H, Nakajima K, Ochiai T, Koide Y, Okazumi SI, Matsubara H, Takeda A, Miyazawa Y, Arima M, Isono K. Detection of serum p53 antibodies in patients with esophageal squamous cell carcinoma: correlation with clinicopathologic features and tumor markers. Oncol Rep. 1998;5:871-874.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
53.  Sobti RC, Parashar K. A study on p53 protein and anti-p53 antibodies in the sera of patients with oesophageal cancer. Mutat Res. 1998;422:271-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
54.  Cawley HM, Meltzer SJ, De Benedetti VM, Hollstein MC, Muehlbauer KR, Liang L, Bennett WP, Souza RF, Greenwald BD, Cottrell J, Salabes A, Bartsch H, Trivers GE. Anti-p53 antibodies in patients with Barrett's esophagus or esophageal carcinoma can predate cancer diagnosis. Gastroenterology. 1998;115:19-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
55.  Jin Y, Guan S, Liu L, Sun S, Lee KH, Wei J. Anti-p16 autoantibodies may be a useful biomarker for early diagnosis of esophageal cancer. Asia Pac J Clin Oncol. 2015;11:e37-e41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
56.  Xiu Y, Sun B, Jiang Y, Wang A, Liu L, Liu Y, Sun S, Huangfu M. Diagnostic Value of the Survivin Autoantibody in Four Types of Malignancies. Genet Test Mol Biomarkers. 2018;22:384-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
57.  Oshima Y, Shimada H, Yajima S, Nanami T, Matsushita K, Nomura F, Kainuma O, Takiguchi N, Soda H, Ueda T, Iizasa T, Yamamoto N, Yamamoto H, Nagata M, Yokoi S, Tagawa M, Ohtsuka S, Kuwajima A, Murakami A, Kaneko H. NY-ESO-1 autoantibody as a tumor-specific biomarker for esophageal cancer: screening in 1969 patients with various cancers. J Gastroenterol. 2016;51:30-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 48]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
58.  Fujita S, Wada H, Jungbluth AA, Sato S, Nakata T, Noguchi Y, Doki Y, Yasui M, Sugita Y, Yasuda T, Yano M, Ono T, Chen YT, Higashiyama M, Gnjatic S, Old LJ, Nakayama E, Monden M. NY-ESO-1 expression and immunogenicity in esophageal cancer. Clin Cancer Res. 2004;10:6551-6558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
59.  Zhang J, Wang K, Zhang J, Liu SS, Dai L, Zhang JY. Using proteomic approach to identify tumor-associated proteins as biomarkers in human esophageal squamous cell carcinoma. J Proteome Res. 2011;10:2863-2872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 113]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
60.  Fujita Y, Nakanishi T, Miyamoto Y, Hiramatsu M, Mabuchi H, Miyamoto A, Shimizu A, Takubo T, Tanigawa N. Proteomics-based identification of autoantibody against heat shock protein 70 as a diagnostic marker in esophageal squamous cell carcinoma. Cancer Lett. 2008;263:280-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
61.  Zhang H, Xia J, Wang K, Zhang J. Serum autoantibodies in the early detection of esophageal cancer: a systematic review. Tumour Biol. 2015;36:95-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 3.6]  [Reference Citation Analysis (1)]
62.  Werner S, Chen H, Tao S, Brenner H. Systematic review: serum autoantibodies in the early detection of gastric cancer. Int J Cancer. 2015;136:2243-2252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 55]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
63.  Klade CS, Voss T, Krystek E, Ahorn H, Zatloukal K, Pummer K, Adolf GR. Identification of tumor antigens in renal cell carcinoma by serological proteome analysis. Proteomics. 2001;1:890-898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
64.  Kijanka G, Murphy D. Protein arrays as tools for serum autoantibody marker discovery in cancer. J Proteomics. 2009;72:936-944.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 70]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
65.  Kim T, Grobmyer SR, Smith R, Ben-David K, Ang D, Vogel SB, Hochwald SN. Esophageal cancer--the five year survivors. J Surg Oncol. 2011;103:179-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 98]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
66.  Pepe MS, Feng Z, Janes H, Bossuyt PM, Potter JD. Pivotal evaluation of the accuracy of a biomarker used for classification or prediction: standards for study design. J Natl Cancer Inst. 2008;100:1432-1438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 540]  [Cited by in F6Publishing: 505]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
67.  Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6:857-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5705]  [Cited by in F6Publishing: 5849]  [Article Influence: 324.9]  [Reference Citation Analysis (0)]
68.  Lujambio A, Lowe SW. The microcosmos of cancer. Nature. 2012;482:347-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 802]  [Cited by in F6Publishing: 859]  [Article Influence: 71.6]  [Reference Citation Analysis (0)]
69.  Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K, Rassenti L, Kipps T, Negrini M, Bullrich F, Croce CM. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99:15524-15529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3675]  [Cited by in F6Publishing: 3636]  [Article Influence: 165.3]  [Reference Citation Analysis (0)]
70.  Wang J, Sen S. MicroRNA functional network in pancreatic cancer: from biology to biomarkers of disease. J Biosci. 2011;36:481-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 51]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
71.  Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105:10513-10518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5636]  [Cited by in F6Publishing: 6046]  [Article Influence: 377.9]  [Reference Citation Analysis (0)]
72.  Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, Li Q, Li X, Wang W, Zhang Y, Wang J, Jiang X, Xiang Y, Xu C, Zheng P, Zhang J, Li R, Zhang H, Shang X, Gong T, Ning G, Wang J, Zen K, Zhang J, Zhang CY. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18:997-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3218]  [Cited by in F6Publishing: 3383]  [Article Influence: 211.4]  [Reference Citation Analysis (0)]
73.  Zhang C, Wang C, Chen X, Yang C, Li K, Wang J, Dai J, Hu Z, Zhou X, Chen L, Zhang Y, Li Y, Qiu H, Xing J, Liang Z, Ren B, Yang C, Zen K, Zhang CY. Expression profile of microRNAs in serum: a fingerprint for esophageal squamous cell carcinoma. Clin Chem. 2010;56:1871-1879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 251]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
74.  Wan J, Wu W, Che Y, Kang N, Zhang R. Insights into the potential use of microRNAs as a novel class of biomarkers in esophageal cancer. Dis Esophagus. 2016;29:412-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
75.  Yao C, Liu HN, Wu H, Chen YJ, Li Y, Fang Y, Shen XZ, Liu TT. Diagnostic and Prognostic Value of Circulating MicroRNAs for Esophageal Squamous Cell Carcinoma: a Systematic Review and Meta-analysis. J Cancer. 2018;9:2876-2884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
76.  Wang K, Chen D, Meng Y, Xu J, Zhang Q. Clinical evaluation of 4 types of microRNA in serum as biomarkers of esophageal squamous cell carcinoma. Oncol Lett. 2018;16:1196-1204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
77.  Sharma P, Saraya A, Sharma R. Serum-based six-miRNA signature as a potential marker for EC diagnosis: Comparison with TCGA miRNAseq dataset and identification of miRNA-mRNA target pairs by integrated analysis of TCGA miRNAseq and RNAseq datasets. Asia Pac J Clin Oncol. 2018;14:e289-e301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
78.  Zhang L, Dong B, Ren P, Ye H, Shi J, Qin J, Wang K, Wang P, Zhang J. Circulating plasma microRNAs in the detection of esophageal squamous cell carcinoma. Oncol Lett. 2018;16:3303-3318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
79.  Lv H, He Z, Wang H, Du T, Pang Z. Differential expression of miR-21 and miR-75 in esophageal carcinoma patients and its clinical implication. Am J Transl Res. 2016;8:3288-3298.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Li BX, Yu Q, Shi ZL, Li P, Fu S. Circulating microRNAs in esophageal squamous cell carcinoma: association with locoregional staging and survival. Int J Clin Exp Med. 2015;8:7241-7250.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Ye M, Ye P, Zhang W, Rao J, Xie Z. [Diagnostic values of salivary versus and plasma microRNA-21 for early esophageal cancer]. Nan Fang Yi Ke Da Xue Xue Bao. 2014;34:885-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
82.  Kurashige J, Kamohara H, Watanabe M, Tanaka Y, Kinoshita K, Saito S, Hiyoshi Y, Iwatsuki M, Baba Y, Baba H. Serum microRNA-21 is a novel biomarker in patients with esophageal squamous cell carcinoma. J Surg Oncol. 2012;106:188-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 81]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
83.  Wang B, Zhang Q. The expression and clinical significance of circulating microRNA-21 in serum of five solid tumors. J Cancer Res Clin Oncol. 2012;138:1659-1666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 150]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
84.  Komatsu S, Ichikawa D, Takeshita H, Tsujiura M, Morimura R, Nagata H, Kosuga T, Iitaka D, Konishi H, Shiozaki A, Fujiwara H, Okamoto K, Otsuji E. Circulating microRNAs in plasma of patients with oesophageal squamous cell carcinoma. Br J Cancer. 2011;105:104-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 199]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
85.  Zhou X, Wen W, Zhu J, Huang Z, Zhang L, Zhang H, Qi LW, Shan X, Wang T, Cheng W, Zhu D, Yin Y, Chen Y, Zhu W, Shu Y, Liu P. A six-microRNA signature in plasma was identified as a potential biomarker in diagnosis of esophageal squamous cell carcinoma. Oncotarget. 2017;8:34468-34480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 42]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
86.  Wu C, Li M, Hu C, Duan H. Clinical significance of serum miR-223, miR-25 and miR-375 in patients with esophageal squamous cell carcinoma. Mol Biol Rep. 2014;41:1257-1266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 65]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
87.  Wu C, Wang C, Guan X, Liu Y, Li D, Zhou X, Zhang Y, Chen X, Wang J, Zen K, Zhang CY, Zhang C. Diagnostic and prognostic implications of a serum miRNA panel in oesophageal squamous cell carcinoma. PLoS One. 2014;9:e92292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 85]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
88.  Komatsu S, Ichikawa D, Hirajima S, Kawaguchi T, Miyamae M, Okajima W, Ohashi T, Arita T, Konishi H, Shiozaki A, Fujiwara H, Okamoto K, Yagi N, Otsuji E. Plasma microRNA profiles: identification of miR-25 as a novel diagnostic and monitoring biomarker in oesophageal squamous cell carcinoma. Br J Cancer. 2014;111:1614-1624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
89.  Zhang T, Zhao D, Wang Q, Yu X, Cui Y, Guo L, Lu SH. MicroRNA-1322 regulates ECRG2 allele specifically and acts as a potential biomarker in patients with esophageal squamous cell carcinoma. Mol Carcinog. 2013;52:581-590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
90.  Sudo K, Kato K, Matsuzaki J, Boku N, Abe S, Saito Y, Daiko H, Takizawa S, Aoki Y, Sakamoto H, Niida S, Takeshita F, Fukuda T, Ochiya T. Development and Validation of an Esophageal Squamous Cell Carcinoma Detection Model by Large-Scale MicroRNA Profiling. JAMA Netw Open. 2019;2:e194573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
91.  Li M, Wu F, Ji Y, Yang L, Li F. Meta-analysis of microRNAs as potential biomarkers for detecting esophageal carcinoma in Asian populations. Int J Biol Markers. 2017;32:e375-e383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
92.  Rosenfeld N, Aharonov R, Meiri E, Rosenwald S, Spector Y, Zepeniuk M, Benjamin H, Shabes N, Tabak S, Levy A, Lebanony D, Goren Y, Silberschein E, Targan N, Ben-Ari A, Gilad S, Sion-Vardy N, Tobar A, Feinmesser M, Kharenko O, Nativ O, Nass D, Perelman M, Yosepovich A, Shalmon B, Polak-Charcon S, Fridman E, Avniel A, Bentwich I, Bentwich Z, Cohen D, Chajut A, Barshack I. MicroRNAs accurately identify cancer tissue origin. Nat Biotechnol. 2008;26:462-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 728]  [Cited by in F6Publishing: 698]  [Article Influence: 43.6]  [Reference Citation Analysis (0)]
93.  Mattick JS. Non-coding RNAs: the architects of eukaryotic complexity. EMBO Rep. 2001;2:986-991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 582]  [Cited by in F6Publishing: 530]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
94.  Harrow J, Frankish A, Gonzalez JM, Tapanari E, Diekhans M, Kokocinski F, Aken BL, Barrell D, Zadissa A, Searle S, Barnes I, Bignell A, Boychenko V, Hunt T, Kay M, Mukherjee G, Rajan J, Despacio-Reyes G, Saunders G, Steward C, Harte R, Lin M, Howald C, Tanzer A, Derrien T, Chrast J, Walters N, Balasubramanian S, Pei B, Tress M, Rodriguez JM, Ezkurdia I, van Baren J, Brent M, Haussler D, Kellis M, Valencia A, Reymond A, Gerstein M, Guigó R, Hubbard TJ. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 2012;22:1760-1774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3435]  [Cited by in F6Publishing: 3205]  [Article Influence: 291.4]  [Reference Citation Analysis (0)]
95.  Alam T, Medvedeva YA, Jia H, Brown JB, Lipovich L, Bajic VB. Promoter analysis reveals globally differential regulation of human long non-coding RNA and protein-coding genes. PLoS One. 2014;9:e109443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 67]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
96.  Huarte M. The emerging role of lncRNAs in cancer. Nat Med. 2015;21:1253-1261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1625]  [Cited by in F6Publishing: 1915]  [Article Influence: 239.4]  [Reference Citation Analysis (0)]
97.  Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, Wang Y, Brzoska P, Kong B, Li R, West RB, van de Vijver MJ, Sukumar S, Chang HY. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464:1071-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3723]  [Cited by in F6Publishing: 4052]  [Article Influence: 289.4]  [Reference Citation Analysis (0)]
98.  Lv XB, Lian GY, Wang HR, Song E, Yao H, Wang MH. Long noncoding RNA HOTAIR is a prognostic marker for esophageal squamous cell carcinoma progression and survival. PLoS One. 2013;8:e63516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 131]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
99.  Yarmishyn AA, Kurochkin IV. Long noncoding RNAs: a potential novel class of cancer biomarkers. Front Genet. 2015;6:145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 198]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
100.  Huang X, Yuan T, Tschannen M, Sun Z, Jacob H, Du M, Liang M, Dittmar RL, Liu Y, Liang M, Kohli M, Thibodeau SN, Boardman L, Wang L. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genomics. 2013;14:319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 765]  [Cited by in F6Publishing: 741]  [Article Influence: 67.4]  [Reference Citation Analysis (0)]
101.  Arita T, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Shoda K, Kawaguchi T, Hirajima S, Nagata H, Kubota T, Fujiwara H, Okamoto K, Otsuji E. Circulating long non-coding RNAs in plasma of patients with gastric cancer. Anticancer Res. 2013;33:3185-3193.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Hu HB, Jie HY, Zheng XX. Three Circulating LncRNA Predict Early Progress of Esophageal Squamous Cell Carcinoma. Cell Physiol Biochem. 2016;40:117-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
103.  Tong YS, Wang XW, Zhou XL, Liu ZH, Yang TX, Shi WH, Xie HW, Lv J, Wu QQ, Cao XF. Identification of the long non-coding RNA POU3F3 in plasma as a novel biomarker for diagnosis of esophageal squamous cell carcinoma. Mol Cancer. 2015;14:3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 238]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
104.  Wang W, He X, Zheng Z, Ma X, Hu X, Wu D, Wang M. Serum HOTAIR as a novel diagnostic biomarker for esophageal squamous cell carcinoma. Mol Cancer. 2017;16:75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 91]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
105.  Zhihua Z, Weiwei W, Lihua N, Jianying Z, Jiang G. p53-induced long non-coding RNA PGM5-AS1 inhibits the progression of esophageal squamous cell carcinoma through regulating miR-466/PTEN axis. IUBMB Life. 2019;71:1492-1502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
106.  Shi T, Gao G, Cao Y. Long Noncoding RNAs as Novel Biomarkers Have a Promising Future in Cancer Diagnostics. Dis Markers. 2016;2016:9085195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 177]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
107.  Mandel P, Metais P. Les acides nucléiques du plasma sanguin chez l'homme. C R Seances Soc Biol Fil. 1948;142:241-243.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977;37:646-650.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11:426-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1954]  [Cited by in F6Publishing: 1989]  [Article Influence: 153.0]  [Reference Citation Analysis (0)]
110.  Caldas C, Hahn SA, Hruban RH, Redston MS, Yeo CJ, Kern SE. Detection of K-ras mutations in the stool of patients with pancreatic adenocarcinoma and pancreatic ductal hyperplasia. Cancer Res. 1994;54:3568-3573.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, Yao SL. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol Biomarkers Prev. 1994;3:67-71.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta. 2007;1775:181-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 418]  [Article Influence: 23.2]  [Reference Citation Analysis (0)]
113.  Luo H, Li H, Hu Z, Wu H, Liu C, Li Y, Zhang X, Lin P, Hou Q, Ding G, Wang Y, Li S, Wei D, Qiu F, Li Y, Wu S. Noninvasive diagnosis and monitoring of mutations by deep sequencing of circulating tumor DNA in esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 2016;471:596-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
114.  Spindler KL, Pallisgaard N, Andersen RF, Brandslund I, Jakobsen A. Circulating free DNA as biomarker and source for mutation detection in metastatic colorectal cancer. PLoS One. 2015;10:e0108247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 94]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
115.  Kawakami K, Brabender J, Lord RV, Groshen S, Greenwald BD, Krasna MJ, Yin J, Fleisher AS, Abraham JM, Beer DG, Sidransky D, Huss HT, Demeester TR, Eads C, Laird PW, Ilson DH, Kelsen DP, Harpole D, Moore MB, Danenberg KD, Danenberg PV, Meltzer SJ. Hypermethylated APC DNA in plasma and prognosis of patients with esophageal adenocarcinoma. J Natl Cancer Inst. 2000;92:1805-1811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 249]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
116.  Hibi K, Taguchi M, Nakayama H, Takase T, Kasai Y, Ito K, Akiyama S, Nakao A. Molecular detection of p16 promoter methylation in the serum of patients with esophageal squamous cell carcinoma. Clin Cancer Res. 2001;7:3135-3138.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Liu JB, Qiang FL, Dong J, Cai J, Zhou SH, Shi MX, Chen KP, Hu ZB. Plasma DNA methylation of Wnt antagonists predicts recurrence of esophageal squamous cell carcinoma. World J Gastroenterol. 2011;17:4917-4921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
118.  Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, Foye A, Lloyd P, Nykter M, Beer TM, Alumkal JJ, Thomas GV, Reiter RE, Rettig MB, Evans CP, Gao AC, Chi KN, Small EJ, Gleave ME. Concordance of Circulating Tumor DNA and Matched Metastatic Tissue Biopsy in Prostate Cancer. J Natl Cancer Inst. 2017;109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 256]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
119.  Beaver JA, Jelovac D, Balukrishna S, Cochran R, Croessmann S, Zabransky DJ, Wong HY, Toro PV, Cidado J, Blair BG, Chu D, Burns T, Higgins MJ, Stearns V, Jacobs L, Habibi M, Lange J, Hurley PJ, Lauring J, VanDenBerg D, Kessler J, Jeter S, Samuels ML, Maar D, Cope L, Cimino-Mathews A, Argani P, Wolff AC, Park BH. Detection of cancer DNA in plasma of patients with early-stage breast cancer. Clin Cancer Res. 2014;20:2643-2650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 286]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
120.  Lebofsky R, Decraene C, Bernard V, Kamal M, Blin A, Leroy Q, Rio Frio T, Pierron G, Callens C, Bieche I, Saliou A, Madic J, Rouleau E, Bidard FC, Lantz O, Stern MH, Le Tourneau C, Pierga JY. Circulating tumor DNA as a non-invasive substitute to metastasis biopsy for tumor genotyping and personalized medicine in a prospective trial across all tumor types. Mol Oncol. 2015;9:783-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 209]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
121.  Frenel JS, Carreira S, Goodall J, Roda D, Perez-Lopez R, Tunariu N, Riisnaes R, Miranda S, Figueiredo I, Nava-Rodrigues D, Smith A, Leux C, Garcia-Murillas I, Ferraldeschi R, Lorente D, Mateo J, Ong M, Yap TA, Banerji U, Gasi Tandefelt D, Turner N, Attard G, de Bono JS. Serial Next-Generation Sequencing of Circulating Cell-Free DNA Evaluating Tumor Clone Response To Molecularly Targeted Drug Administration. Clin Cancer Res. 2015;21:4586-4596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 160]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
122.  Esposito A, Criscitiello C, Locatelli M, Milano M, Curigliano G. Liquid biopsies for solid tumors: Understanding tumor heterogeneity and real time monitoring of early resistance to targeted therapies. Pharmacol Ther. 2016;157:120-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
123.  André F, Bachelot T, Commo F, Campone M, Arnedos M, Dieras V, Lacroix-Triki M, Lacroix L, Cohen P, Gentien D, Adélaide J, Dalenc F, Goncalves A, Levy C, Ferrero JM, Bonneterre J, Lefeuvre C, Jimenez M, Filleron T, Bonnefoi H. Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol. 2014;15:267-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 295]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
124.  Levy M, Benesova L, Lipska L, Belsanova B, Minarikova P, Veprekova G, Zavoral M, Minarik M. Utility of cell-free tumour DNA for post-surgical follow-up of colorectal cancer patients. Anticancer Res. 2012;32:1621-1626.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Reinert T, Schøler LV, Thomsen R, Tobiasen H, Vang S, Nordentoft I, Lamy P, Kannerup AS, Mortensen FV, Stribolt K, Hamilton-Dutoit S, Nielsen HJ, Laurberg S, Pallisgaard N, Pedersen JS, Ørntoft TF, Andersen CL. Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery. Gut. 2016;65:625-634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 314]  [Article Influence: 39.3]  [Reference Citation Analysis (0)]
126.  Nicholson JK, Lindon JC, Holmes E. 'Metabonomics': understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data. Xenobiotica. 1999;29:1181-1189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2918]  [Cited by in F6Publishing: 2605]  [Article Influence: 104.2]  [Reference Citation Analysis (0)]
127.  De Preter V, Verbeke K. Metabolomics as a diagnostic tool in gastroenterology. World J Gastrointest Pharmacol Ther. 2013;4:97-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
128.  Armitage EG, Barbas C. Metabolomics in cancer biomarker discovery: current trends and future perspectives. J Pharm Biomed Anal. 2014;87:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 234]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
129.  Ma H, Hasim A, Mamtimin B, Kong B, Zhang HP, Sheyhidin I. Plasma free amino acid profiling of esophageal cancer using high-performance liquid chromatography spectroscopy. World J Gastroenterol. 2014;20:8653-8659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
130.  Puchades-Carrasco L, Pineda-Lucena A. Metabolomics Applications in Precision Medicine: An Oncological Perspective. Curr Top Med Chem. 2017;17:2740-2751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 63]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
131.  Wishart DS. Is Cancer a Genetic Disease or a Metabolic Disease? EBioMedicine. 2015;2:478-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 89]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
132.  Jin H, Qiao F, Chen L, Lu C, Xu L, Gao X. Serum metabolomic signatures of lymph node metastasis of esophageal squamous cell carcinoma. J Proteome Res. 2014;13:4091-4103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 65]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
133.  Mir SA, Rajagopalan P, Jain AP, Khan AA, Datta KK, Mohan SV, Lateef SS, Sahasrabuddhe N, Somani BL, Keshava Prasad TS, Chatterjee A, Veerendra Kumar KV, VijayaKumar M, Kumar RV, Gundimeda S, Pandey A, Gowda H. LC-MS-based serum metabolomic analysis reveals dysregulation of phosphatidylcholines in esophageal squamous cell carcinoma. J Proteomics. 2015;127:96-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
134.  Liu R, Peng Y, Li X, Wang Y, Pan E, Guo W, Pu Y, Yin L. Identification of plasma metabolomic profiling for diagnosis of esophageal squamous-cell carcinoma using an UPLC/TOF/MS platform. Int J Mol Sci. 2013;14:8899-8911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
135.  Lee M, Rhee I. Cytokine Signaling in Tumor Progression. Immune Netw. 2017;17:214-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
136.  Spangler JB, Moraga I, Mendoza JL, Garcia KC. Insights into cytokine-receptor interactions from cytokine engineering. Annu Rev Immunol. 2015;33:139-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 170]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
137.  Wilson J, Balkwill F. The role of cytokines in the epithelial cancer microenvironment. Semin Cancer Biol. 2002;12:113-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 122]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
138.  Serefoglou Z, Yapijakis C, Nkenke E, Vairaktaris E. Genetic association of cytokine DNA polymorphisms with head and neck cancer. Oral Oncol. 2008;44:1093-1099.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
139.  Kozłowski M, Laudański W, Mroczko B, Szmitkowski M, Milewski R, Łapuć G. Serum tissue inhibitor of metalloproteinase 1 (TIMP-1) and vascular endothelial growth factor A (VEGF-A) are associated with prognosis in esophageal cancer patients. Adv Med Sci. 2013;58:227-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
140.  Krzystek-Korpacka M, Matusiewicz M, Diakowska D, Grabowski K, Blachut K, Konieczny D, Kustrzeba-Wojcicka I, Terlecki G, Banas T. Elevation of circulating interleukin-8 is related to lymph node and distant metastases in esophageal squamous cell carcinomas--implication for clinical evaluation of cancer patient. Cytokine. 2008;41:232-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
141.  Krzystek-Korpacka M, Matusiewicz M, Diakowska D, Grabowski K, Blachut K, Banas T. Up-regulation of VEGF-C secreted by cancer cells and not VEGF-A correlates with clinical evaluation of lymph node metastasis in esophageal squamous cell carcinoma (ESCC). Cancer Lett. 2007;249:171-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
142.  Ren Y, Law S, Huang X, Lee PY, Bacher M, Srivastava G, Wong J. Macrophage migration inhibitory factor stimulates angiogenic factor expression and correlates with differentiation and lymph node status in patients with esophageal squamous cell carcinoma. Ann Surg. 2005;242:55-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 60]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
143.  Shimada H, Takeda A, Nabeya Y, Okazumi SI, Matsubara H, Funami Y, Hayashi H, Gunji Y, Kobayashi S, Suzuki T, Ochiai T. Clinical significance of serum vascular endothelial growth factor in esophageal squamous cell carcinoma. Cancer. 2001;92:663-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
144.  Kozlowski M, Kowalczuk O, Milewski R, Chyczewski L, Niklinski J, Laudański J. Serum vascular endothelial growth factors C and D in patients with oesophageal cancer. Eur J Cardiothorac Surg. 2010;38:260-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
145.  Tong Q, Wang XL, Li SB, Yang GL, Jin S, Gao ZY, Liu XB. Combined detection of IL-6 and IL-8 is beneficial to the diagnosis of early stage esophageal squamous cell cancer: a preliminary study based on the screening of serum markers using protein chips. Onco Targets Ther. 2018;11:5777-5787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
146.  Ren Y, Cao B, Law S, Xie Y, Lee PY, Cheung L, Chen Y, Huang X, Chan HM, Zhao P, Luk J, Vande Woude G, Wong J. Hepatocyte growth factor promotes cancer cell migration and angiogenic factors expression: a prognostic marker of human esophageal squamous cell carcinomas. Clin Cancer Res. 2005;11:6190-6197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 118]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
147.  Łukaszewicz-Zając M, Mroczko B, Kozłowski M, Nikliński J, Laudański J, Szmitkowski M. Higher importance of interleukin 6 than classic tumor markers (carcinoembryonic antigen and squamous cell cancer antigen) in the diagnosis of esophageal cancer patients. Dis Esophagus. 2012;25:242-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]